TW201711687A - Solid dosage forms of palbociclib - Google Patents

Solid dosage forms of palbociclib Download PDF

Info

Publication number
TW201711687A
TW201711687A TW105117213A TW105117213A TW201711687A TW 201711687 A TW201711687 A TW 201711687A TW 105117213 A TW105117213 A TW 105117213A TW 105117213 A TW105117213 A TW 105117213A TW 201711687 A TW201711687 A TW 201711687A
Authority
TW
Taiwan
Prior art keywords
dosage form
solid dosage
acid
weight
minutes
Prior art date
Application number
TW105117213A
Other languages
Chinese (zh)
Other versions
TWI635863B (en
Inventor
法迪 艾貝辛
馬修 穆拉尼
雷巍 山克
巴巴拉 史邦
堅 王
Original Assignee
輝瑞股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=56092955&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=TW201711687(A) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by 輝瑞股份有限公司 filed Critical 輝瑞股份有限公司
Publication of TW201711687A publication Critical patent/TW201711687A/en
Application granted granted Critical
Publication of TWI635863B publication Critical patent/TWI635863B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/194Carboxylic acids, e.g. valproic acid having two or more carboxyl groups, e.g. succinic, maleic or phthalic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1652Polysaccharides, e.g. alginate, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2086Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2095Tabletting processes; Dosage units made by direct compression of powders or specially processed granules, by eliminating solvents, by melt-extrusion, by injection molding, by 3D printing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Inorganic Chemistry (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The present invention relates to solid dosage forms of palbociclib comprising a water-soluble acid. The dosage forms described herein have desirable pharmacokinetic characteristics.

Description

帕博西里之固態劑型 Parbsili solid dosage form

本發明關於6-乙醯基-8-環戊基-5-甲基-2-(5-哌-1-基-吡啶-2-基胺基)-8H-吡啶并[2,3-d]嘧啶-7-酮(下文的帕博西里)之固態劑型,其具有展現有利的貯存穩定性及溶解性質之所欲藥物動力學特徵。 The present invention relates to 6-ethylindenyl-8-cyclopentyl-5-methyl-2-(5-piperidin a solid dosage form of 1-yl-pyridin-2-ylamino)-8H-pyrido[2,3-d]pyrimidin-7-one (P.B.), which exhibits favorable storage stability and The desired pharmacokinetic profile of the solubility properties.

帕博西里為CDK4及CDK6之有效力及選擇性抑制劑,其具有以下結構: Pabsili is a potent and selective inhibitor of CDK4 and CDK6, which has the following structure:

帕博西里說明於WHO Drug Information,Vol.27,No.2,page 172(2013)中。帕博西里及其醫藥上可接受之鹽揭示於國際公開案號WO 2003/062236及美國專利案號 6,936,612、7,208,489和7,456,168;國際公開案號WO 2005/005426及美國專利案號7,345,171和7,863,278;國際公開案號WO 2008/032157及美國專利案號7,781,583;及國際公開案號WO 2014/128588中。將每一前述參考文獻的揭示內容以彼之全文併入本文以供參考全文。 Pabsiri is described in WHO Drug Information, Vol. 27, No. 2, page 172 (2013). Pabsiri and its pharmaceutically acceptable salts are disclosed in International Publication No. WO 2003/062236 and U.S. Patent No. 6, 936, 612, 7, 208, 489 and 7, 456, 168; International Publication No. WO 2005/005426 and U.S. Patent Nos. 7,345,171 and 7,863,278; International Publication No. WO 2008/032157, and U.S. Patent No. 7,781,583; and International Publication No. WO 2014/128588. The disclosure of each of the aforementioned references is incorporated herein by reference in its entirety.

帕博西里在美國經核准與來曲唑(letrozole)組合作為初始內分泌療法或在以內分泌療法的疾病進展之後與氟維司群(fulvestrant)組合用於治療激素受體(HR)-陽性、人表皮生長因子2(HER2)-陰性晚期或轉移性乳癌。藥物係由Pfizer以商品名IBRANCE®銷售,其呈包含帕博西里作為游離鹼之經口投予的即時釋放型(IR)膠囊形式。 Pabsiri is approved in the United States in combination with letrozole as an initial endocrine therapy or in combination with fulvestrant for the treatment of hormone receptor (HR)-positive, human after disease progression with endocrine therapy Epidermal growth factor 2 (HER2) - negative advanced or metastatic breast cancer. The drug is marketed by Pfizer under the trade name IBRANCE® in the form of an immediate release (IR) capsule containing oral administration of paclocil as the free base.

帕博西里為二鹼性化合物且具有兩個約7.3(第二哌氮)和4.1(吡啶氮)之pKa的鹼性基團。帕博西里游離鹼之溶解度具有pH依賴性。帕博西里在低pH(2.1-4.5)下為水溶性,且溶解度在pH上升大於4.5時戲劇性地下降。帕博西里在pH 7.9下具有差的水溶解度(9微克/毫升)。伴隨投予之增加胃pH的劑可改變帕博西里游離鹼調配物的溶解度及吸收。 Pabbori is a dibasic compound and has two 7.3 (second piper) A basic group of pKa of nitrogen) and 4.1 (pyridinium). The solubility of the paclitaxel free base is pH dependent. Pabsili is water soluble at low pH (2.1-4.5) and the solubility drops dramatically as the pH rises above 4.5. Pabsiri had poor water solubility (9 μg/ml) at pH 7.9. The agent that increases the pH of the stomach with administration can alter the solubility and absorption of the paclitaxel free base formulation.

當經口給藥時,治療劑之吸收及生物利用率可受許多因素影響,包括受試者是否在進食或空腹狀態及使用某些藥品,諸如質子泵抑制劑(PPI)或H2受體拮抗劑,以及某些醫學症狀。具有pH依賴性溶解度之化合物(特別為鹼性化合物)可展現非所欲藥物動力學性質,諸如差的吸 收及/或降低的生物利用率,其可導致顯著的患者間和患者本身變異性。 When administered orally, the absorption and bioavailability of the therapeutic agent can be affected by a number of factors, including whether the subject is eating or fasting and using certain drugs, such as proton pump inhibitor (PPI) or H2 receptor antagonism. Agents, as well as certain medical symptoms. Compounds with pH-dependent solubility (especially basic compounds) can exhibit undesired pharmacokinetic properties, such as poor absorption Achieving and/or reduced bioavailability can result in significant inter-patient and patient variability.

對發現具有利的溶解及藥物動力學輪廓的改進之帕博西里劑型仍有需求,該改進之劑型亦展示良好的貯存穩定性。吾等驚訝地發現根據本發明的固態劑型展示極佳的貯存穩定性且提供實質上無pH依賴性之帕博西里遞輸,沒有顯著的食物效應或與PPI不利的交互作用。 There is still a need for an improved Pabsiri dosage form found to have a favorable dissolution and pharmacokinetic profile, and the improved dosage form also exhibits good storage stability. We have surprisingly found that the solid dosage form according to the present invention exhibits excellent storage stability and provides substantially pH-independent pabbice delivery without significant food effects or adverse interactions with PPI.

本發明概述 Summary of the invention

在第一態樣中,本發明提供包含帕博西里、水溶性酸及醫藥上可接受之載劑的固態劑型。 In a first aspect, the invention provides a solid dosage form comprising a paclitaxel, a water soluble acid, and a pharmaceutically acceptable carrier.

在第二態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中當劑型添加至具有於50rpm自旋之葉片的標準USP 2旋轉葉片裝置中包含37℃下500毫升10mM pH 5.5之乙酸鹽緩衝液的試驗介質中時,其溶解:(a)在15分鐘內不少於35%之帕博西里;(b)在30分鐘內不少於45%之帕博西里;(c)在60分鐘內不少於55%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 In a second aspect, the invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form is added to a standard USP 2 rotating blade device having a blade of spin at 50 rpm comprising 500 at 37 °C In the test medium of 10 mM acetate buffer pH 5.5, it dissolves: (a) not less than 35% of Pabori in 15 minutes; (b) not less than 45% of Pabbo in 30 minutes Siri; (c) not less than 55% of Pabori in 60 minutes; or (d) two or more of (a), (b) and (c).

在第三態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中當劑型添加至具有於50rpm自旋之葉片的標準USP 2旋轉葉片裝置中包含37℃下500毫升50mM pH 6.5之磷酸鹽緩衝液及0.1M NaCl的試驗介 質中時,其溶解:(a)在15分鐘內不少於15%之帕博西里;(b)在30分鐘內不少於20%之帕博西里;(c)在60分鐘內不少於25%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 In a third aspect, the invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form is added to a standard USP 2 rotary vane device having a blade of spin at 50 rpm comprising 500 at 37 °C Test of milliliter 50mM phosphate buffer pH 6.5 and 0.1M NaCl In the middle, it dissolves: (a) less than 15% of Pabori in 15 minutes; (b) less than 20% of Pabori in 30 minutes; (c) in 60 minutes In 25% of Pabbori; or (d) (a), (b) and (c) two or more.

在一些實施態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中當劑型添加至具有於50rpm自旋之葉片的標準USP 2旋轉葉片裝置中包含37℃下500毫升50mM pH 6.5之磷酸鹽緩衝液及0.1M NaCl的試驗介質中時,其溶解:(a)在15分鐘內不少於20%之帕博西里;(b)在30分鐘內不少於30%之帕博西里;(c)在60分鐘內不少於25%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 In some embodiments, the present invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form is added to a standard USP 2 rotating blade device having a blade of spin at 50 rpm comprising 500 at 37 °C When immersed in 50 mM phosphate buffer solution of pH 6.5 and 0.1 M NaCl in the test medium, it dissolves: (a) not less than 20% of Pabori in 15 minutes; (b) not less than 30 in 30 minutes % of Pabsiri; (c) not less than 25% of Pabori in 60 minutes; or (d) two or more of (a), (b) and (c).

在其他的實施態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中當劑型添加至具有於50rpm自旋之葉片的標準USP 2旋轉葉片裝置中包含37℃下500毫升50mM pH 6.5之磷酸鹽緩衝液及0.1M NaCl的試驗介質中時,其溶解(a)在15分鐘內不少於40%之帕博西里;(b)在30分鐘內不少於35%之帕博西里;(c)在60分鐘內不少於25%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 In other embodiments, the present invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form is added to a standard USP 2 rotating blade device having a blade of spin at 50 rpm at 37 ° C. 500 ml of 50 mM phosphate buffer of pH 6.5 and 0.1 M NaCl in the test medium, which dissolves (a) not less than 40% of Pabsiri in 15 minutes; (b) not less than 35 in 30 minutes % of Pabsiri; (c) not less than 25% of Pabori in 60 minutes; or (d) two or more of (a), (b) and (c).

在第四態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中劑型:(a)在投予受試者單一經口劑量之後在血漿濃度相對時間曲線下面積(AUC)具有從約0.8至約1.25的平均進食/空腹比;(b)在投予受試 者單一經口劑量之後具有從約0.8至約1.25之最大血漿濃度(Cmax)的平均進食/空腹比;或(c)(a)與(b)二者。 In a fourth aspect, the invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form: (a) the area under the plasma concentration versus time curve after administration of the subject to a single oral dose (AUC) has an average fed/fasting ratio of from about 0.8 to about 1.25; (b) an average of eating from a maximum oral plasma concentration ( Cmax ) of from about 0.8 to about 1.25 after administration of a single oral dose to the subject. Fasting ratio; or (c) both (a) and (b).

在第五態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中劑型:(a)在投予受試者單一經口劑量之後提供含有等效量的帕博西里之對照用即時釋放型(IR)經口膠囊的80%至125%之平均空腹AUC的範圍內之平均空腹AUC;或(b)在投予受試者單一經口劑量之後提供含有等效量的帕博西里之對照用即時釋放型(IR)經口膠囊的80%至125%之平均空腹Cmax的範圍內之平均空腹Cmax;或(c)(a)與(b)二者。 In a fifth aspect, the invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form: (a) provides an equivalent amount of Pabo after administration of a single oral dose to the subject Siri control is an average fasting AUC within the range of 80% to 125% of the average fasting AUC of immediate release (IR) oral capsules; or (b) providing equivalent after administration of a single oral dose to the subject Pabo Xi in the amount of control with immediate-release (IR) by an average of 80 to 125% of the opening of the capsule fasting average of the range of fasting plasma C max C max; or (c) (a) and both (b) .

在第六態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中劑型:(a)在投予受試者單一經口劑量之後提供在質子泵抑制劑(PPI)存在下的平均AUC係在沒有PPI存在下的平均AUC之80%至125%的範圍內;(b)在投予受試者單一經口劑量之後提供在質子泵抑制劑(PPI)存在下的平均Cmax係在沒有PPI存在下的平均Cmax之80%至125%的範圍內;或(c)(a)與(b)二者。在一些此等實施態樣中,PPI為瑞貝拉唑(rabeprazole)。 In a sixth aspect, the invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form: (a) is provided in a proton pump inhibitor (PPI) after administration of a single oral dose to the subject The average AUC in the presence of 80% to 125% of the mean AUC in the absence of PPI; (b) provided in the presence of a proton pump inhibitor (PPI) after administration of a single oral dose to the subject in the range of 80 to 125% of the average C max of the mean C max is no system in the presence of a PPI; or (c) (a) (b ) and both. In some such embodiments, the PPI is rabeprazole.

在第七態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中劑型在30℃及75%相對濕度(RH)下貯存96天之後展現少於0.3重量%之酸加成物。 In a seventh aspect, the invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form exhibits less than 0.3% by weight after storage for 96 days at 30 ° C and 75% relative humidity (RH) Acid addition.

在第八態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中劑型在30℃及75% RH下貯存2年之後展現少於1.0重量%之酸加成物。 In an eighth aspect, the present invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form exhibits less than 1.0% by weight acid adduct after storage for 2 years at 30 ° C and 75% RH .

在第九態樣中,本發明提供本文所述之實施態樣中任一者之固態劑型,其中劑型在25℃/60% RH下貯存1年之後展現少於0.05重量%之酸加成物。在一些此等實施態樣中,劑型係與乾燥劑盒使用熱感應密封包裝在瓶內。 In a ninth aspect, the invention provides a solid dosage form of any of the embodiments described herein, wherein the dosage form exhibits less than 0.05% by weight acid adduct after storage for one year at 25 ° C / 60% RH . In some such embodiments, the dosage form and the desiccant cartridge are packaged in a bottle using a heat sensitive seal.

在本發明的態樣中之各者的一些實施態樣中,活性醫藥成分(API)帕博西里構成劑型的從約10%重量%至約35重量%。在特定的實施態樣中,帕博西里構成劑型的約20重量%。 In some embodiments of each of the aspects of the invention, the active pharmaceutical ingredient (API) Pabburry comprises from about 10% by weight to about 35% by weight of the dosage form. In a particular embodiment, the Pabsili constitutes about 20% by weight of the dosage form.

在本發明的態樣中之各者的一些實施態樣中,水溶性酸構成劑型的從約5重量%至約40重量%。在特別的實施態樣中,水溶性酸構成劑型的從約5重量%至約25重量%。在其他的實施態樣中,水溶性酸構成劑型的從約5重量%至約15重量%。在更特別的實施態樣中,水溶性酸構成劑型的約10重量%。 In some embodiments of each of the aspects of the invention, the water soluble acid comprises from about 5% to about 40% by weight of the dosage form. In a particular embodiment, the water soluble acid comprises from about 5% to about 25% by weight of the dosage form. In other embodiments, the water soluble acid comprises from about 5% to about 15% by weight of the dosage form. In a more particular embodiment, the water soluble acid comprises about 10% by weight of the dosage form.

在一些此等實施態樣中,水溶性酸係選自由琥珀酸、蘋果酸和酒石酸所組成之群組。在特定的實施態樣中,水溶性酸為琥珀酸。在其他的實施態樣中,水溶性酸為蘋果酸。在另外的實施態樣中,水溶性酸為酒石酸。 In some such embodiments, the water soluble acid is selected from the group consisting of succinic acid, malic acid, and tartaric acid. In a particular embodiment, the water soluble acid is succinic acid. In other embodiments, the water soluble acid is malic acid. In another embodiment, the water soluble acid is tartaric acid.

在本文所述態樣中之各者的較佳實施態樣中,固態劑型包含從約10重量%至約35重量%之帕博西里、從約5重量%至約25重量%之選自由琥珀酸、蘋果酸和酒石酸所 組成之群組的水溶性酸及醫藥上可接受之載劑。在特別的實施態樣中,水溶性酸為琥珀酸。在一些此等實施態樣中,固態劑型包含約20重量%之帕博西里、約10重量%之琥珀酸及醫藥上可接受之載劑。 In a preferred embodiment of each of the aspects described herein, the solid dosage form comprises from about 10% to about 35% by weight of Pabbori, from about 5% to about 25% by weight selected from the group consisting of amber Acid, malic acid and tartaric acid A group of water soluble acids and pharmaceutically acceptable carriers. In a particular embodiment, the water soluble acid is succinic acid. In some such embodiments, the solid dosage form comprises about 20% by weight of Pabsiri, about 10% by weight of succinic acid, and a pharmaceutically acceptable carrier.

在本文所述態樣中之各者的一些實施態樣中,醫藥上可接受之載劑包含下列醫藥上可接受之賦形劑中之一或多者:稀釋劑、崩解劑、結合劑、潤滑劑、助滑劑及表面活性劑。此等賦形劑可以顆粒內或顆粒外方式併入錠劑型式中,且錠劑可包含相同或不同的賦形劑作為顆粒內或顆粒外組份。例如,錠劑調配物可包含可相同或不同的顆粒內潤滑劑、顆粒外潤滑劑或顆粒內和顆粒外潤滑劑二者。 In some embodiments of each of the aspects described herein, the pharmaceutically acceptable carrier comprises one or more of the following pharmaceutically acceptable excipients: diluent, disintegrant, binder , lubricants, slip agents and surfactants. Such excipients may be incorporated into the tablet form either intragranularly or extragranularly, and the lozenge may comprise the same or different excipients as either intragranular or extragranular components. For example, the lozenge formulation can comprise both the same or different intragranular lubricants, extragranular lubricants or both intragranular and extragranular lubricants.

在本文所述態樣中之各者的一些實施態樣中,醫藥上可接受之載劑包含至少一種稀釋劑,其中稀釋劑構成固態劑型的約50重量%至約75重量%。在特定的實施態樣中,載劑包含至少一種選自由下列所組成之群組的稀釋劑:微晶纖維素、乳糖單水合物、甘露醇、山梨醇、木糖醇、碳酸鎂、磷酸氫鈣(dibasic calcium phosphate)和磷酸鈣(tribasic calcium phosphate)。在特定的實施態樣中,稀釋劑為微晶纖維素。在一些此等實施態樣中,稀釋劑為微晶纖維素。 In some embodiments of each of the aspects described herein, the pharmaceutically acceptable carrier comprises at least one diluent, wherein the diluent comprises from about 50% to about 75% by weight of the solid dosage form. In a particular embodiment, the carrier comprises at least one diluent selected from the group consisting of microcrystalline cellulose, lactose monohydrate, mannitol, sorbitol, xylitol, magnesium carbonate, hydrogen phosphate Dibasic calcium phosphate and tribasic calcium phosphate. In a particular embodiment, the diluent is microcrystalline cellulose. In some such embodiments, the diluent is microcrystalline cellulose.

在本文所述態樣中之各者的其他實施態樣中,醫藥上可接受之載劑包含潤滑劑,其中潤滑劑構成固態劑型的從約0.5重量%至約10重量%。在特定的實施態樣中,載劑包含至少一種選自由下列所組成之群組的潤滑劑:硬脂酸 鎂、硬脂酸鈣、硬脂酸鋅和硬脂基富馬酸鈉。在特定的實施態樣中,潤滑劑為硬脂酸鎂,其可包括於顆粒內、顆粒外或二者。在其他的實施態樣中,潤滑劑為硬脂基富馬酸鈉。其他的實施態樣包含硬脂酸鎂及硬脂基富馬酸鈉二者作為潤滑劑,其可包括於顆粒內、顆粒外或二者。 In other embodiments of each of the aspects described herein, the pharmaceutically acceptable carrier comprises a lubricant, wherein the lubricant comprises from about 0.5% to about 10% by weight of the solid dosage form. In a particular embodiment, the carrier comprises at least one lubricant selected from the group consisting of stearic acid Magnesium, calcium stearate, zinc stearate and sodium stearyl fumarate. In a particular embodiment, the lubricant is magnesium stearate, which may be included in the particles, out of the particles, or both. In other embodiments, the lubricant is sodium stearyl fumarate. Other embodiments include both magnesium stearate and sodium stearyl fumarate as lubricants, which may be included in the particles, out of the particles, or both.

在本文所述態樣中之各者的另外實施態樣中,醫藥上可接受之載劑包含至少一種崩解劑,其中崩解劑構成固態劑型的從約5重量%至約10重量%。在特定的實施態樣中,載劑包含至少一種選自由下列所組成之群組的崩解劑:交聯聚維酮(crospovidone)、交聯羧甲基纖維素鈉(croscarmellose sodium)和乙醇酸澱粉鈉(sodium starch glycolate)。在特定的實施態樣中,崩解劑為交聯聚維酮。 In a further embodiment of each of the aspects described herein, the pharmaceutically acceptable carrier comprises at least one disintegrant, wherein the disintegrant constitutes from about 5% by weight to about 10% by weight of the solid dosage form. In a particular embodiment, the carrier comprises at least one disintegrant selected from the group consisting of crospovidone, croscarmellose sodium, and glycolic acid. Sodium starch glycolate. In a particular embodiment, the disintegrant is crospovidone.

在本文所述態樣中之各者常見的實施態樣中,本發明的固態劑型係呈錠劑形式。在一些實施態樣中,錠劑經包膜。在一些實施態樣中,錠劑為單層錠劑。在其他的實施態樣中,錠劑為雙層錠劑。在特別的實施態樣中,本發明的錠劑包含25毫克、75毫克、100毫克或125毫克量之帕博西里。在特定的實施態樣中,本發明的錠劑包含125毫克量之帕博西里。 In a common embodiment of each of the aspects described herein, the solid dosage form of the invention is in the form of a tablet. In some embodiments, the tablet is coated. In some embodiments, the tablet is a single layer tablet. In other embodiments, the lozenge is a bilayer tablet. In a particular embodiment, the lozenge of the present invention comprises Pabsiri in an amount of 25 mg, 75 mg, 100 mg or 125 mg. In a particular embodiment, the tablet of the present invention comprises a 125 mg amount of Pabbori.

在本發明態樣中之各者常見的實施態樣中,固態劑型係呈錠劑形式。在一些實施態樣中,錠劑為單層錠劑。在其他的實施態樣中,錠劑為雙層錠劑。 In a common embodiment of each of the aspects of the invention, the solid dosage form is in the form of a tablet. In some embodiments, the tablet is a single layer tablet. In other embodiments, the lozenge is a bilayer tablet.

在本文所述態及實施態樣的一些實施態樣中,在劑型 中的帕博西里之量為25毫克、75毫克、100毫克或125毫克。在特定的實施態樣中,在劑型中的帕博西里之量為125毫克。 In some embodiments of the states and embodiments described herein, in a dosage form The amount of Pabsili in the case is 25 mg, 75 mg, 100 mg or 125 mg. In a particular embodiment, the amount of Pabbori in the dosage form is 125 mg.

本文所述之本發明的實施態樣中之各者可與本文所述之本發明的一或多個實施態樣組合,其與所組合之實施態樣不一致。 Each of the embodiments of the invention described herein may be combined with one or more embodiments of the invention described herein, which are inconsistent with the embodiments.

圖1. 包含八種試驗水溶性酸(蘋果酸、順丁烯二酸、琥珀酸、反丁烯二酸、酒石酸、甲苯磺酸、苯甲酸及苯磺酸)之原型調配物在具有於50rpm自旋之葉片的USP 2裝置中在37℃下於pH 5.5之10mM乙酸鈉緩衝溶液中的試管內溶解數據。 Figure 1. Prototype formulation containing eight experimental water-soluble acids (malic acid, maleic acid, succinic acid, fumaric acid, tartaric acid, toluenesulfonic acid, benzoic acid, and benzenesulfonic acid) at 50 rpm The spin-leafed USP 2 apparatus was lysed in a test tube at 37 ° C in a 10 mM sodium acetate buffer solution at pH 5.5.

圖2. 包含琥珀酸、蘋果酸和酒石酸之原型調配物在具有於50rpm自旋之葉片的USP 2裝置中在37℃下於pH 5.5之10mM乙酸鈉緩衝溶液中的試管內溶解數據。 Figure 2. In vitro in vitro dissolution data for a prototype formulation containing succinic acid, malic acid, and tartaric acid in a USP 2 device with blades at 50 rpm spin at 37 ° C in 10 mM sodium acetate buffer at pH 5.5.

圖3. 包含琥珀酸之原型調配物在具有於50rpm自旋之葉片的USP 2裝置中在37℃下於含有+0.1M NaCl的pH 6.5之50mM磷酸鹽緩衝溶液中的非漏槽(non-sink)試管內溶解數據。 Figure 3. Non-leakage (non-) in a 50 mM phosphate buffer solution containing +0.1 M NaCl at pH 6.5 in a USP 2 unit with blades at 50 rpm spin at 37 ° C in a prototype formulation containing succinic acid. Sink) Dissolve data in the tube.

圖4. 調配物A1、A2及B在具有於50rpm自旋之葉片的USP 2裝置中在37℃下於含有+0.1M NaCl的pH 6.5之50mM磷酸鹽緩衝液中的非漏槽試管內溶解數據。 Figure 4. Formulations A1, A2, and B were dissolved in a non-slotted tube in a 50 mM phosphate buffer pH 6.5 containing +0.1 M NaCl at 37 °C in a USP 2 device with 50 rpm spin blades. data.

本發明可藉由參考下列詳細說明的本發明較佳的實施態樣及其中所包括的實施例而更輕易地瞭解。應瞭解本文所使用的術語僅以說明特定的實施態樣為目的,並不意欲為限制。應進一步瞭解本文所使用的術語係以相關技術中已知的其傳統意義而給出,除非本文經具體的定義。 The present invention may be more readily understood by reference to the preferred embodiments of the invention described hereinbelow. It is understood that the terminology used herein is for the purpose of illustration and description It is to be further understood that the terms used herein are given in their conventional sense as known in the related art, unless specifically defined herein.

如本文所使用的單數形式〝一(a)〞、〝一(an)〞及〝該(the)〞包括複數個指示物,除非另有其他指示。例如,〝一〞賦形劑包括一或多個賦形劑。 As used herein, the singular forms "a", "an", "the" and "the" are meant to include the plural unless otherwise indicated. For example, an excipient includes one or more excipients.

術語〝約〞意指有一值落在由一般熟習本技術領域者認為可接受的平均誤差標準內。術語〝約〞時常係指其述及之值或範圍的±15%,較佳為±10%,及更佳為±5%。例如,〝約10重量%〞意指10重量%±1.5重量%,較佳為10重量%±1重量%,及更佳為10重量%±0.5重量%。 The term "about" means that a value falls within the standard of error that is generally accepted by those skilled in the art. The term "about" is used to mean ±15%, preferably ±10%, and more preferably ±5%, of the value or range recited. For example, about 10% by weight of 〞 means 10% by weight ± 1.5% by weight, preferably 10% by weight ± 1% by weight, and more preferably 10% by weight ± 0.5% by weight.

除非另有其他指示,帕博西里係指6-乙醯基-8-環戊基-5-甲基-2-(5-哌-1-基-吡啶-2-胺基)-8H-吡啶并[2,3-d]嘧啶-7-酮之游離鹼,其可以晶形或非晶形,或非晶形與晶形之混合存在。 Unless otherwise indicated, Paporicil refers to 6-acetamido-8-cyclopentyl-5-methyl-2-(5-piperidin The free base of 1-yl-pyridin-2-amino)-8H-pyrido[2,3-d]pyrimidin-7-one, which may be crystalline or amorphous, or a mixture of amorphous and crystalline forms.

經口投予之藥物的吸收可受到藥物通過胃腸(GI)道時的pH變化之影響。吸收可發生在沿著GI道的不同位置上,例如在臉頰內側或在胃、十二指腸、空腸、迴腸或結腸中。在各吸收位置上的pH不同,胃的pH(pH 1-3.5)與小腸的pH(pH 4.5-8)有顯著的差別。當藥物通 過GI道時,具有pH依賴性溶解度之藥物可自溶液沉澱,導致在劑量或患者之間的吸收程度及/或吸收率的患者間或患者本身變異性。 The absorption of the drug administered orally can be affected by the pH change of the drug as it passes through the gastrointestinal (GI) tract. Absorption can occur at different locations along the GI tract, such as on the inside of the cheek or in the stomach, duodenum, jejunum, ileum, or colon. The pH at each absorption site is different, and the pH of the stomach (pH 1-3.5) is significantly different from the pH of the small intestine (pH 4.5-8). When drugs pass Over the GI tract, drugs with pH-dependent solubility can precipitate from solution, resulting in a degree of absorption between the dose or patient and/or the rate of absorption between patients or the patient itself.

GI道之pH亦可建基於患者或受試者是否為進食或空腹狀態而改變。藥物的胃滯留時間通常在食物的存在下比在空腹狀態更長。若藥物的生物利用率受到存在或不存在於GI道中的食物而有顯著的影響時,則稱藥物展現〝食物效應〞。胃排空率亦可影響在沿著GI道的不同位置上有效吸收之溶液中的藥物濃度。 The pH of the GI tract can also be varied based on whether the patient or subject is in a fed or fasting state. The gastric retention time of the drug is usually longer in the presence of food than in the fasting state. If the bioavailability of the drug is significantly affected by the presence or absence of food in the GI tract, the drug is said to exhibit a licking effect. Gastric emptying rate can also affect the concentration of drug in a solution that is effectively absorbed at different locations along the GI tract.

共同投予之某些藥品以及醫學症狀(諸如無胃酸症)亦可影響GI道之pH。使用降酸劑(諸如質子泵抑制劑(PPI)或H2受體拮抗劑)可導致相對高的胃pH,其可導致具有pH依賴性溶解度之藥物僅部分溶解於胃中。未溶解之藥物的進一步溶解可由於上腸道較高的pH環境中的低溶解度而受到抑制。這可導致具有pH依賴性溶解度之藥物的不均勻溶解,增加藥物-藥物交互作用的風險且可能造成降低吸收及降低治療效益。 Certain drugs and medical symptoms (such as no acidosis) that are co-administered may also affect the pH of the GI tract. The use of an acid reducing agent, such as a proton pump inhibitor (PPI) or an H2 receptor antagonist, can result in a relatively high gastric pH, which can result in a drug having pH dependent solubility only partially soluble in the stomach. Further dissolution of the undissolved drug can be inhibited due to the low solubility in the upper pH environment of the upper intestinal tract. This can result in uneven solubilization of the drug with pH-dependent solubility, increasing the risk of drug-drug interaction and possibly resulting in reduced absorption and reduced therapeutic benefit.

在健康的志願者中的研究顯示暴露於帕博西里治療(以游離鹼膠囊每天投予一次125毫克)以進食受試者(AUCinf,23%-27%;Cmax,21%-24%)相對空腹受試者(AUCinf,39%;Cmax;73%)而略微增加且大大降低在進食狀態下的PK變異性。Ruiz-Garcia等人之Annals of Oncology(2014)25(suppl_4):iv146-iv164.10.1093/annonc/mdu331。因為在進食狀態中觀察到降低的 患者間變異性,所以在美國包裝插頁上建議市售帕博西里游離鹼膠囊與食物一起服用。 Studies in healthy volunteers showed exposure to paclicilil (125 mg administered daily in free base capsules) to eat subjects (AUCinf, 23%-27%; Cmax, 21%-24%) relative Fasting subjects (AUCinf, 39%; Cmax; 73%) slightly increased and greatly reduced PK variability in the fed state. Ruiz-Garcia et al., Annals of Oncology (2014) 25 (suppl_4): iv 146-iv 164.10.1093/annonc/mdu331. Because a decrease was observed in the fed state Inter-patient variability, so it is recommended to take the commercially available Pabsiri free base capsules with food on the US package insert.

當化合物調配成錠劑或其他固態劑型時,希望發展在高於那些典型遇到的溫度及相對濕度水平下具有貯存穩定性的調配物。亦可尋求其他希望的調配物性質,諸如快速溶解,使得錠劑迅速溶解且使藥物有效吸收。因此,良好的貯存穩定性及快速溶解尤其是作為本發明的所欲特徵而尋求的特性。 When the compounds are formulated into tablets or other solid dosage forms, it is desirable to develop formulations that have storage stability above those typically encountered temperatures and relative humidity levels. Other desirable formulation properties may also be sought, such as rapid dissolution, such that the tablet dissolves rapidly and the drug is effectively absorbed. Therefore, good storage stability and rapid dissolution are especially desirable properties as a desirable feature of the present invention.

藥物溶解代表影響全身性吸收率的關鍵因素。已發展出各種試管內方法以評定醫藥調配物之溶解性質,且溶解測試有時被用作為直接評估藥物生物利用率的替代法。參見例如Emmanuel等人之Pharmaceutics(2010),2:351-363且引入本文以供參考。溶解測試測量在控制的測試條件下經限定的時期自藥物產品(亦即錠劑或膠囊)釋出且溶解在溶解介質中的API百分比。為了維持漏槽狀態(sink condition),在溶解介質中的藥物飽溶解度應為至少三倍藥物濃度。低溶解度化合物的溶解有時可在非漏槽狀態下測定。溶解受到API性質(例如粒徑、晶形、裝填密度)、藥物產品組成(例如載藥量、賦形劑)、製造方法(例如壓縮力)及在貯存條件下的穩定性(例如溫度、濕度)的影響。 Drug dissolution represents a key factor affecting systemic absorption. Various in vitro methods have been developed to assess the solubility properties of pharmaceutical formulations, and dissolution testing is sometimes used as an alternative to directly assessing drug bioavailability. See, for example, Pharmaceutics (2010), Emmanuel et al., 2: 351-363, incorporated herein by reference. The dissolution test measures the percentage of API that is released from the drug product (ie, lozenge or capsule) and dissolved in the dissolution medium for a defined period of time under controlled test conditions. In order to maintain a sink condition, the drug saturation in the dissolution medium should be at least three times the drug concentration. The dissolution of low solubility compounds can sometimes be measured in a non-leaky state. Dissolution is affected by API properties (eg particle size, crystal form, packing density), pharmaceutical product composition (eg drug loading, excipients), manufacturing methods (eg compression forces) and stability under storage conditions (eg temperature, humidity) Impact.

在加速老化條件下評定固態劑型的化學貯存穩定性之方法已說明於文獻中。參見例如S.T.Colgan,T.J.Watson,R.D.Whipple,R.Nosal,J.V.Beaman,D.De Antonis之“The Application of Science and Risk Based Concepts to Drug Substance Stability Strategies”J.Pharm.Innov.7:205-2013(2012);Waterman KC,Carella AJ,Gumkowski MJ等人之Improved protocol and data analysis for accelerated shelf-life estimation of solid dosage forms.Pharm Res 2007;24(4):780-90;及S.T.Colgan,R.J.Timpano,D.Diaz,M.Roberts,R.Weaver,K.Ryan,K.Fields,G.Scrivens之“Opportunities for Lean Stability Strategies”J.Pharm.Innov.9:259-271(2014)。 Methods for assessing the chemical storage stability of solid dosage forms under accelerated aging conditions are described in the literature. See, for example, S.T. Colgan, T.J. Watson, R.D. Whipple, R. Nosal, J.V. Beaman, D.De Antonis's "The Application of Science and Risk Based Concepts to Drug Substance Stability Strategies" J. Pharm. Innov. 7: 205-2013 (2012); Waterman KC, Carella AJ, Gumkowski MJ et al., Improved protocol and data analysis for Accelerated Shelf-life estimation of solid dosage forms. Pharm Res 2007;24(4):780-90; and STColgan, RJTimpano, D.Diaz, M.Roberts, R.Weaver, K.Ryan, K. Fields, G "Opportunities for Lean Stability Strategies" by Scrivens, J. Pharm. Innov. 9:259-271 (2014).

本發明的固態劑型之帕博西里的哌基部分及水溶性酸的酸加成物形成為經監控以評定貯存穩定性之重要降解物。 The solid dosage form of the present invention, Pabsiri's piperazine The base moiety and the acid adduct of the water soluble acid form an important degradant that is monitored to assess storage stability.

如本文進一步說明,本發明提供包含帕博西里、水溶性酸及醫藥上可接受之載劑的固態劑型及彼之製造和使用方法。 As further described herein, the present invention provides solid dosage forms comprising paclicil, a water soluble acid, and a pharmaceutically acceptable carrier, and methods of making and using same.

固態劑型包括但不限於即時釋放型錠劑及膠囊、控制釋放型(CR)錠劑及膠囊、速溶劑型、可咀嚼劑型、囊袋等等。本發明的劑型較佳地呈錠劑形式,包括單層或雙層錠劑。 Solid dosage forms include, but are not limited to, immediate release tablets and capsules, controlled release (CR) tablets and capsules, fast solvent type, chewable dosage forms, pouches and the like. The dosage form of the invention is preferably in the form of a tablet, including a single layer or a bilayer tablet.

本發明的〝固態劑型〞為安全用於經口投予人類的醫藥上可接受之固態劑型,其中在劑型中的所有賦形劑為醫藥上可接受於經口調配物中使用,換言之,其安全用於人類攝取。在常見的實施態樣中,固態劑型為錠劑。 The bismuth solid dosage form of the present invention is a pharmaceutically acceptable solid dosage form for safe oral administration to humans, wherein all excipients in the dosage form are pharmaceutically acceptable for use in oral formulations, in other words, Safe for human ingestion. In a common embodiment, the solid dosage form is a lozenge.

如本文所使用的術語〝單位劑量(unit dose或unit dosage)〞係指含有經計算以產生所欲治療效應之預定量的活性成分之物理分立單位。單位劑量可呈錠劑、膠囊、囊袋等等形式,在本文稱為〝單位劑型〞。 The term unit dose or unit as used herein. Dosage refers to a physically discrete unit containing a predetermined amount of the active ingredient calculated to produce the desired therapeutic effect. The unit dosage can be in the form of a tablet, capsule, sachet, or the like, which is referred to herein as a unit dosage form.

如本文所使用的術語〝空腹〞經定義如下:限定在至少10小時(亦即10小時)隔夜空腹之後(其中發生0卡路里攝入)的給藥狀態。受試者可以240毫升水投予劑型。應在給藥後至少4小時不容許進食。除了在藥物投予之前及之後1小時以外,可容許依需要飲水。 The term "empty belly" as used herein is defined as follows: limited to at least 10 hours (ie, 10 hours) The state of administration after an overnight fasting (where 0 calorie intake occurs). Subjects can be administered a dosage form of 240 ml of water. Eating should not be allowed for at least 4 hours after administration. Drinking water as needed may be allowed, except 1 hour before and after the drug is administered.

如本文所使用的術語〝進食〞經定義如下:限定在至少10小時隔夜空腹之後(其中發生0卡路里攝入),接著受試者開始建議之膳食的給藥狀態。受試者應在30分鐘或之內進餐;然而藥物產品應在開始進餐之後30分鐘投予。藥物產品可以240毫升水投予。應在給藥後至少4小時不容許進食。除了在藥物投予之前及之後1小時以外,可容許依需要飲水。 The term "ingestion" as used herein is defined as follows: after at least 10 hours of overnight fasting (where 0 calorie intake occurs), the subject then begins the recommended state of administration of the meal. Subjects should eat within 30 minutes; however, the drug product should be administered 30 minutes after the start of the meal. The drug product can be administered in 240 ml of water. Eating should not be allowed for at least 4 hours after administration. Drinking water as needed may be allowed, except 1 hour before and after the drug is administered.

為了評定進食/空腹比,可投予單一經口劑量之帕博西里:在高脂肪、高卡路里膳食(~800-1000卡路里,具有分別來自蛋白質、碳水化合物和脂肪的150、250及500-600卡路里)之後30分鐘;在低脂肪、低卡路里膳食(~400-500卡路里,具有分別來自蛋白質、碳水化合物和脂肪的120、250及28-35卡路里)之後30分鐘;或介於適當的脂肪及卡路里含量膳食(由15%蛋白質、50%碳水化合物及35%脂肪所組成的~500-700卡路里)的膳食之間(在之後1小時/在之前2小時)。 To assess the feeding/fasting ratio, a single oral dose of Pabsiri can be administered: in a high-fat, high-calorie diet (~800-1000 calories, with 150, 250, and 500-600 from protein, carbohydrate, and fat, respectively) 30 minutes after calories; 30 minutes after a low-fat, low-calorie diet (~400-500 calories with 120, 250, and 28-35 calories from protein, carbohydrate, and fat, respectively); or between proper fat and A meal of calorie-rich meal (~500-700 calories consisting of 15% protein, 50% carbohydrates, and 35% fat) (1 hour later / 2 hours before).

高脂肪及高卡路里膳食可用作為進食狀態下的試驗膳食。高脂肪試驗膳食的實例會有兩個以奶油煎的雞蛋、兩條培根、兩片有奶油的土司、四盎司炸薯餅及八盎司全乳。 High fat and high calorie meals are available as a test meal in the fed state. Examples of high fat test meals are two milk-fried eggs, two bacon, two creamy toasts, four ounces of potato wedges, and eight ounces of whole milk.

在平均血清濃度相對時間曲線下面積(AUC)之計算為醫藥技術中熟知的程序且說明於例如Welling,"Pharmacokinetics Processes and Mathematics," ACS Monograph 185(1986)中。如本文所使用的AUC包括在單一劑量之後自時間0外推至無限時間之濃度-時間曲線下的面積或在穩定態/多重劑量之後自時間0至給藥間隔結束時間之濃度-時間曲線下的面積。 The calculation of the area under the mean serum concentration versus time curve (AUC) is a procedure well known in the medical arts and is described, for example, in Welling, "Pharmacokinetics Processes and Mathematics," ACS Monograph 185 (1986). AUC as used herein includes the area under the concentration-time curve extrapolated from time 0 to infinity after a single dose or under a concentration-time curve from time 0 to the end of the dosing interval after steady state/multiple doses Area.

另外,Cmax、Cmin,ss、Tmax及清除半衰期(t½)之計算亦為一般熟習本技術領域者已知且說明於例如Shargel,Wu-Pong,and Yu之Applied Biopharmaceutics and Pharmacokinetics(2005)中。 In addition, the calculation of C max , C min, ss , T max and the elimination half-life (t1⁄2) is also known to those skilled in the art and is described, for example, in Shargel, Wu-Pong, and Yu, Applied Biopharmaceutics and Pharmacokinetics (2005). in.

為了測定平均進食/空腹比,先計算帕博西里在進食狀態下的血漿濃度相對時間曲線下的平均面積(例如AUC0-inf)對帕博西里在空腹狀態下的血漿濃度相對時間曲線下的平均面積(例如AUC0-inf)之個別比,且接著將對應的個別比一起平均。在此方式中,其為經測定之各個對應的個別比之平均。 In order to determine the average feeding/fasting ratio, the average area under the plasma concentration versus time curve of the pascaliri in the fed state (for example, AUC 0-inf ) was calculated for the plasma concentration versus time curve of the pasoriberi in the fasting state. The individual ratios of the average areas (e.g., AUC 0-inf ) are then averaged together for the corresponding individual ratios. In this manner, it is the average of the respective individual ratios determined.

質子泵抑制劑(PPI)為降低胃酸產生之熟知的藥物類別,藉此修改胃pH。代表性PPI包括例如瑞貝拉唑、奧美拉唑(omeprazole)(包括S-和B-形式,Na和Mg 鹽)、蘭索拉唑(lansoprazole)、泮托拉唑(pantoprazole)、埃索美拉唑(eseprazole)及類似者。 Proton pump inhibitors (PPIs) are well-known classes of drugs that reduce gastric acid production, thereby modifying gastric pH. Representative PPIs include, for example, rabeprazole, omeprazole (including S- and B-forms, Na and Mg) Salt), lansoprazole, pantoprazole, eseprazole and the like.

如本文所使用的術語〝對照用即時釋放型(IR)經口膠囊〞係指如實施例11中所述之市售帕博西里IR膠囊調配物。缺少水溶性酸,但實質上在其他方面與實施例1之調配物相同的此調配物與羥乙磺酸鹽調配物(ISE)及游離鹼錠劑劑型可一起於本文稱為對照物。 As used herein, the term "immediate release type (IR) oral capsule" refers to a commercially available Pabsiri IR capsule formulation as described in Example 11. This formulation lacking a water soluble acid, but otherwise substantially identical to the formulation of Example 1, can be referred to herein as a control together with the isethionate formulation (ISE) and the free base lozenge dosage form.

〝溶解試驗1〞係指帕博西里劑型的下列試驗。溶解試驗係在如美國藥典(USP)溶解試驗章節(Dissolution Test Chapter)711,裝置2中所揭示之標準的USP 2旋轉葉片裝置中進行。葉片係以50rpm旋轉且將劑型添加至37℃下500毫升10mM pH 5.5之乙酸鹽緩衝液中。在試驗開始(例如劑型插入裝置中)之後於適當的時間以高性能液相層析術(HPLC)分析來自試驗介質的過濾等分液(通常為1.5毫升)之帕博西里。溶解結果係以測試之帕博西里總劑量相對於時間的溶解百分比記述。 〝 Dissolution test 1 〞 refers to the following test of the Pabsiri dosage form. The dissolution test is carried out in a standard USP 2 rotary vane apparatus as disclosed in the United States Pharmacopoeia (USP) Dissolution Test Chapter 711, apparatus 2. The leaves were spun at 50 rpm and the dosage form was added to 500 ml of 10 mM pH 5.5 acetate buffer at 37 °C. The filtered aliquot (usually 1.5 ml) from the test medium was analyzed by high performance liquid chromatography (HPLC) at the appropriate time after the start of the experiment (eg in the dosage form insertion device). The dissolution results are reported as the percentage of dissolution of the total dose of Pabsiri tested over time.

〝溶解試驗2〞係指帕博西里劑型的下列試驗。溶解試驗係在美國藥典(USP)溶解試驗章節711,裝置2中所揭示之標準的USP 2旋轉葉片裝置中進行。葉片係以50rpm旋轉且將劑型添加至37℃下500毫升50mM pH 6.5之磷酸鹽緩衝液及0.1M NaCl中。在試驗開始(例如劑型插入裝置中)之後於適當的時間以高性能液相層析術(HPLC)分析來自試驗介質的過濾等分液(通常為1.5毫升)之帕博西里。溶解結果係以測試之帕博西里總劑量 相對於時間的溶解百分比記述。 〝 Dissolution test 2 〞 refers to the following test of the Pabsiri dosage form. The dissolution test was carried out in the USP 2 rotary vane apparatus of the standard disclosed in the United States Pharmacopoeia (USP) dissolution test section 711, apparatus 2. The leaves were spun at 50 rpm and the dosage form was added to 500 ml of 50 mM phosphate buffer pH 6.5 and 0.1 M NaCl at 37 °C. The filtered aliquot (usually 1.5 ml) from the test medium was analyzed by high performance liquid chromatography (HPLC) at the appropriate time after the start of the experiment (eg in the dosage form insertion device). The dissolution result was measured by the total dose of Pabsiri tested. The percentage of dissolution relative to time is described.

術語〝乾式粒化〞意指將主體活性產物與至少一種賦形劑摻合之方法。接著將摻合物壓縮或壓實,以形成壓縮材料或〝壓製物〞。此材料可藉由壓碎、研磨或切割而分裂成無水粒狀粒子,以形成顆粒。粒子可隨意地進一步加工。壓碎、研磨或切割方法包含縮減壓縮材料尺寸的操作,諸如藉由磨碎或那些熟習本技術領域者已知的其他操作而完成。 The term "dry granulation" means a method of blending a host active product with at least one excipient. The blend is then compressed or compacted to form a compressed material or crucible compact. This material can be split into anhydrous particulate particles by crushing, grinding or cutting to form granules. The particles can be further processed at will. The crushing, grinding or cutting process involves the operation of reducing the size of the compressed material, such as by grinding or other operations known to those skilled in the art.

在本文所使用與組成物中存在的酸有關的術語〝水溶性〞係指在25℃下於水中具有至少0.2重量%之溶解度的酸。水溶性酸可為有機或無機酸,且較佳為具有至少一個pKa值之有機酸,該值為至少一個(較佳為至少兩個)比在藥物中存在的鹼性基團之最高pKa更小的pK單元。在帕博西里的例子中,水溶性酸具有約4.1及7.3之pKa值,酸較佳地具有小於6.3之pKa,且更佳為小於5.3之pKa。水溶性有機酸包括例如C2-C8或C2-C6脂族單或多羧酸,且較佳為C4-C6脂族單或多羧酸。特別佳為C4-C6二羧酸,其可為飽和或不飽和。 The term "water-soluble oxime" as used herein in connection with the acid present in the composition refers to an acid having a solubility in water of at least 0.2% by weight at 25 °C. The water-soluble acid may be an organic or inorganic acid, and is preferably an organic acid having at least one pKa value, the value being at least one (preferably at least two) higher than the highest pKa of the basic group present in the drug. Small pK unit. In the case of Pabbori, the water soluble acid has a pKa of about 4.1 and 7.3, and the acid preferably has a pKa of less than 6.3, and more preferably a pKa of less than 5.3. The water-soluble organic acid includes, for example, a C 2 -C 8 or C 2 -C 6 aliphatic mono or polycarboxylic acid, and is preferably a C 4 -C 6 aliphatic mono or polycarboxylic acid. Particularly preferred is a C 4 -C 6 dicarboxylic acid which may be saturated or unsaturated.

本發明的固態劑型可包含單一水溶性酸,或可包括二或更多個此等酸之組合。在本發明的選擇性實施態樣中,水溶性酸係選自由琥珀酸、蘋果酸和酒石酸所組成之群組。在本發明特定的較佳實施態樣中,水溶性酸為琥珀酸。 The solid dosage form of the invention may comprise a single water soluble acid or may comprise a combination of two or more such acids. In an alternative embodiment of the invention, the water soluble acid is selected from the group consisting of succinic acid, malic acid and tartaric acid. In a particular preferred embodiment of the invention, the water soluble acid is succinic acid.

可將水溶性酸在粒化前與藥物組合或可與顆粒外賦形 劑一起併入劑型中。在雙層錠劑中,水溶性酸可存在於含有帕博西里之活性層中,併入單獨的酸層中,或水溶性酸類(彼等可相同或不同)可併入活性層及酸層二者中。 The water-soluble acid can be combined with the drug before granulation or can be shaped outside the particle The agents are incorporated together into a dosage form. In the bilayer tablet, the water-soluble acid may be present in the active layer containing pabsoli, incorporated into a separate acid layer, or the water-soluble acid (which may be the same or different) may be incorporated into the active layer and the acid layer. In both.

不想受到理論的束縛,一般認為存在於固態劑型中與藥物緊密接觸之酸係以帕博西里與酸之間的交互作用方式增加溶解。在經口投予受試者之後,本發明的固態劑型由此提供與投予缺少水溶性酸的帕博西里調配物相比而增加溶液中的藥物局部濃度。 Without wishing to be bound by theory, it is believed that the acid present in close contact with the drug in a solid dosage form increases dissolution by the interaction between pabbice and acid. After oral administration to a subject, the solid dosage form of the present invention thus provides for increasing the local concentration of the drug in the solution as compared to administering a Pabsiri formulation lacking the water soluble acid.

在一些實施態樣中,本文所述之實施態樣中之任一者的固態劑型在溶解試驗1的條件下(pH 5.5之乙酸鹽緩衝液,37℃)溶解:(a)在15分鐘內不少於35%之帕博西里;(b)在30分鐘內不少於45%之帕博西里;(c)在60分鐘內不少於55%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 In some embodiments, the solid dosage form of any of the embodiments described herein is dissolved under the conditions of Dissolution Test 1 (pH 5.5 acetate buffer, 37 ° C): (a) within 15 minutes. Not less than 35% of Pabori; (b) not less than 45% of Pabori in 30 minutes; (c) not less than 55% of Pabori in 60 minutes; or (d) (a Two or more of), (b) and (c).

在另外的實施態樣中,本發明的固態劑型在溶解試驗1的條件下溶解:(a)不少於25%、30%、35%、40%、45%或50%或超過50%之帕博西里於15分鐘內溶解;(b)不少於35%、40%、45%、50%、55%或60%或超過60%之帕博西里於30分鐘內溶解;及/或(c)不少於45%、50%、55%、60%、65%或70%或超過70%之帕博西里於60分鐘內溶解。 In another embodiment, the solid dosage form of the invention is dissolved under the conditions of dissolution test 1: (a) not less than 25%, 30%, 35%, 40%, 45% or 50% or more than 50% Pabsili dissolves within 15 minutes; (b) not less than 35%, 40%, 45%, 50%, 55% or 60% or more than 60% of the Pabsili dissolves within 30 minutes; and / or c) Not less than 45%, 50%, 55%, 60%, 65% or 70% or more than 70% of the Pabsili dissolves within 60 minutes.

在一些實施態樣中,本文所述之實施態樣中之任一者的固態劑型在溶解試驗2的非漏槽狀態下(pH 6.5之磷酸鹽緩衝液及0.1M NaCl,37℃)溶解:(a)在15分鐘內 不少於40%之帕博西里;(b)在30分鐘內不少於35%之帕博西里;(c)在60分鐘內不少於25%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 In some embodiments, the solid dosage form of any of the embodiments described herein is dissolved in a non-leaked state of Dissolution Test 2 (pH 6.5 phosphate buffer and 0.1 M NaCl, 37 ° C): (a) within 15 minutes Not less than 40% of Pabsiri; (b) not less than 35% of Pabori in 30 minutes; (c) not less than 25% of Pabori in 60 minutes; or (d) (a Two or more of), (b) and (c).

在其他的實施態樣中,本文所述之實施態樣中之任一者的固態劑型在溶解試驗2的非漏槽狀態下(pH 6.5之磷酸鹽緩衝液及0.1M NaCl,37℃)溶解:(a)在15分鐘內不少於15%之帕博西里;(b)在30分鐘內不少於20%之帕博西里;(c)在60分鐘內不少於25%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 In other embodiments, the solid dosage form of any of the embodiments described herein is dissolved in a non-leaked state of Dissolution Test 2 (pH 6.5 phosphate buffer and 0.1 M NaCl, 37 ° C). : (a) not less than 15% of Pabori in 15 minutes; (b) not less than 20% of Pabori in 30 minutes; (c) not less than 25% of Pabo in 60 minutes Siri; or (d) two or more of (a), (b) and (c).

在其他的實施態樣中,在溶解試驗2下:(a)不少於10%、15%、20%、25%、30%、35%、40%、45%或50%或超過50%之帕博西里於15分鐘內溶解;(b)不少於15%、20%、25%、30%、35%、40%、45%或50%或超過50%之帕博西里於30分鐘內溶解;及/或(c)不少於15%、20%、25%、30%、35%或40%或超過40%之帕博西里於60分鐘內溶解。 In other embodiments, under dissolution test 2: (a) not less than 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50% or more than 50% Pabsili dissolves in 15 minutes; (b) not less than 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50% or more than 50% of Pabbori in 30 minutes Dissolved internally; and/or (c) not less than 15%, 20%, 25%, 30%, 35% or 40% or more than 40% of the Pabsili dissolved within 60 minutes.

在另外的實施態樣中,在溶解試驗2下:(a)不少於30%、35%、40%、45%或50%或超過50%之帕博西里於15分鐘內溶解;(b)不少於25%、30%、35%、40%、45%或50%或超過50%之帕博西里於30分鐘內溶解;及/或(c)不少於15%、20%、25%、30%、35%或40%或超過40%之帕博西里於60分鐘內溶解。 In another embodiment, under dissolution test 2: (a) not less than 30%, 35%, 40%, 45% or 50% or more than 50% of the Pabsili dissolved within 15 minutes; (b ) not less than 25%, 30%, 35%, 40%, 45% or 50% or more than 50% of Pabsili dissolved in 30 minutes; and / or (c) not less than 15%, 20%, 25%, 30%, 35% or 40% or more than 40% of the Pabsili dissolved in 60 minutes.

在一些實施態樣中,本發明提供包含帕博西里、水溶性酸及醫藥上可接受之載劑的固態劑型,其中劑型提供: (a)在投予受試者單一經口劑量之後在血漿濃度相對時間曲線下面積(AUC)從約0.8至約1.25的平均進食/空腹比;(b)在投予受試者單一經口劑量之後從約0.8至約1.25之最大血漿濃度(Cmax)的平均進食/空腹比;或(c)(a)與(b)二者。 In some embodiments, the invention provides a solid dosage form comprising a paclitaxel, a water soluble acid, and a pharmaceutically acceptable carrier, wherein the dosage form provides: (a) in a plasma after administration of a single oral dose to the subject The area under the concentration versus time curve (AUC) is from an average eating/fasting ratio of from about 0.8 to about 1.25; (b) the maximum plasma concentration ( Cmax ) from about 0.8 to about 1.25 after administration of a single oral dose to the subject. Average eating/fasting ratio; or (c) both (a) and (b).

在一些實施態樣中,本發明提供包含帕博西里、水溶性酸及醫藥上可接受之載劑的固態劑型,其中劑型:(a)在投予受試者單一經口劑量之後提供含有等效量的帕博西里之對照用即時釋放型(IR)經口膠囊的80%至125%之平均空腹AUC的範圍內之平均空腹AUC;(b)在投予受試者單一經口劑量之後提供含有等效量的帕博西里之對照用即時釋放型(IR)經口膠囊的80%至125%之平均空腹Cmax的範圍內之平均空腹Cmax;或(c)(a)與(b)二者。 In some embodiments, the invention provides a solid dosage form comprising a paclitaxel, a water soluble acid, and a pharmaceutically acceptable carrier, wherein the dosage form: (a) provides a content after administration of the subject to a single oral dose, etc. Effectiveness of the Pabsiri control average fasting AUC in the range of 80% to 125% of the average fasting AUC of immediate release (IR) oral capsules; (b) after administration of a single oral dose to the subject Pabo Xi provided in the control contained equivalent amounts with immediate-release (IR) by an average of 80 to 125% of the opening of the capsule fasting average of the range of fasting plasma C max C max; or (c) (a) and ( b) Both.

在另外的實施態樣中,本發明提供包含帕博西里、水溶性酸及醫藥上可接受之載劑的固態劑型,其中劑型提供:(a)在投予受試者單一經口劑量之後在質子泵抑制劑(PPI)(較佳為瑞貝拉唑)存在下的平均AUC係在沒有PPI存在下的平均AUC之80%至125%的範圍內;(b)在投予受試者單一經口劑量之後在質子泵抑制劑(PPI)(較佳為瑞貝拉唑)存在下的平均Cmax係在沒有PPI存在下的平均Cmax之80%至125%的範圍內;或(c)(a)與(b)二者。 In a further embodiment, the invention provides a solid dosage form comprising a paclitaxel, a water soluble acid, and a pharmaceutically acceptable carrier, wherein the dosage form provides: (a) after administration of a single oral dose to the subject The average AUC in the presence of a proton pump inhibitor (PPI) (preferably rabeprazole) is in the range of 80% to 125% of the mean AUC in the absence of PPI; (b) in a single subject after oral doses via the proton pump inhibitor (PPI) (preferably azole Ruibei La) present in the system mean C max in the range of 80 to 125% of the average C max of the PPI in the absence of presence; or (c Both (a) and (b).

在其他的實施態樣中,本發明提供包含帕博西里、水 溶性酸及醫藥上可接受之載劑的固態劑型,其中劑型在30℃及75%相對濕度(RH)下貯存96天之後展現少於0.4%、0.35%、0.3%、0.25%、0.2%、0.15%或0.1重量%之酸加成物。 In other embodiments, the present invention provides for inclusion of Pabbori, water a solid dosage form of a soluble acid and a pharmaceutically acceptable carrier, wherein the dosage form exhibits less than 0.4%, 0.35%, 0.3%, 0.25%, 0.2% after storage for 96 days at 30 ° C and 75% relative humidity (RH), 0.15% or 0.1% by weight acid adduct.

在又其他的實施態樣中,本發明提供包含帕博西里、水溶性酸及醫藥上可接受之載劑的固態劑型,其中劑型在30℃及75% RH下貯存2年之後展現少於1.5%、1.4%、1.3%、1.2%、1.1%、1.0%、0.9%、0.8%、0.7%、0.6%、0.5%、0.4%或0.3重量%之酸加成物。 In still other embodiments, the present invention provides a solid dosage form comprising a paclitaxel, a water soluble acid, and a pharmaceutically acceptable carrier, wherein the dosage form exhibits less than 1.5 after storage for 2 years at 30 ° C and 75% RH. %, 1.4%, 1.3%, 1.2%, 1.1%, 1.0%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4% or 0.3% by weight of the acid adduct.

在本發明的態樣及實施態樣中之各者的一些實施態樣中,水溶性酸係選自由琥珀酸、蘋果酸和酒石酸所組成之群組。在本文的態樣及實施態樣中之各者的特定實施態樣中,水溶性酸為琥珀酸。在其他的實施態樣中,水溶性酸為蘋果酸。在另外的實施態樣中,水溶性酸為酒石酸。 In some embodiments of each of the aspects and embodiments of the invention, the water soluble acid is selected from the group consisting of succinic acid, malic acid, and tartaric acid. In a particular embodiment of each of the aspects and embodiments herein, the water soluble acid is succinic acid. In other embodiments, the water soluble acid is malic acid. In another embodiment, the water soluble acid is tartaric acid.

在本發明的態樣中之各者的一些實施態樣中,水溶性酸構成固態劑型的從約5重量%至約40重量%。在特別的實施態樣中,水溶性酸構成固態劑型的從約5重量%至約25重量%。在一些實施態樣中,水溶性酸構成固態劑型的約5重量%、10重量%、15重量%、20重量%、25重量%、30重量%、35重量%或40重量%。 In some embodiments of each of the aspects of the invention, the water soluble acid comprises from about 5% to about 40% by weight of the solid dosage form. In a particular embodiment, the water soluble acid comprises from about 5% to about 25% by weight of the solid dosage form. In some embodiments, the water soluble acid constitutes about 5%, 10%, 15%, 20%, 25%, 30%, 35%, or 40% by weight of the solid dosage form.

在本發明的態樣中之各者的一些實施態樣中,帕博西里構成固態劑型的從約10重量%至約35重量%。在特定的實施態樣中,帕博西里構成固態劑型的約20重量%。在一些實施態樣中,帕博西里構成固態劑型的約5重量 %、10重量%、15重量%、20重量%、25重量%、30重量%、35重量%、40重量%、45重量%或50重量%。 In some embodiments of each of the aspects of the invention, the pablocillin constitutes from about 10% to about 35% by weight of the solid dosage form. In a particular embodiment, the Pabsili constitutes about 20% by weight of the solid dosage form. In some embodiments, the Pabsili constitutes about 5 weights of the solid dosage form. %, 10% by weight, 15% by weight, 20% by weight, 25% by weight, 30% by weight, 35% by weight, 40% by weight, 45% by weight or 50% by weight.

在本文的態樣及實施態樣中之各者的常見實施態樣中,固態劑型為錠劑,較佳為以乾式粒化所形成之錠劑。在一些此等實施態樣中,錠劑為雙層錠劑。在特別的實施態樣中,雙層錠劑包含:(a)包含帕博西里及醫藥上可接受之載劑的活性層;及(b)包含水溶性酸及醫藥上可接受之載劑的酸層。在一些實施態樣中,雙層錠劑包含:(a)包含帕博西里、水溶性酸及醫藥上可接受之載劑的活性層;及(b)包含水溶性酸及醫藥上可接受之載劑的酸層,其中在活性層中的水溶性酸可與在酸層中的水溶性酸相同或不同。在特定的實施態樣中,在活性層中的水溶性酸為琥珀酸及在酸層中的水溶性酸為酒石酸。 In a common embodiment of each of the aspects and embodiments herein, the solid dosage form is a tablet, preferably a tablet formed by dry granulation. In some such embodiments, the lozenge is a bilayer tablet. In a particular embodiment, the bilayer tablet comprises: (a) an active layer comprising paclicil and a pharmaceutically acceptable carrier; and (b) comprising a water soluble acid and a pharmaceutically acceptable carrier Acid layer. In some embodiments, the bilayer tablet comprises: (a) an active layer comprising a Pabsili, a water soluble acid, and a pharmaceutically acceptable carrier; and (b) comprising a water soluble acid and a pharmaceutically acceptable The acid layer of the carrier wherein the water soluble acid in the active layer can be the same or different than the water soluble acid in the acid layer. In a particular embodiment, the water soluble acid in the active layer is succinic acid and the water soluble acid in the acid layer is tartaric acid.

在另一態樣中,本發明提供治療癌症之方法,其包含將治療有效量的本文所述態樣及實施態樣中之任一者的固態劑型投予需要其之受試者。在特別的實施態樣中,癌症為乳癌。在一些此等實施態樣中,乳癌為激素受體陽性(HR+)乳癌。在一些此等實施態樣中,乳癌為雌激素受體陽性(ER+)乳癌。在一些此等實施態樣中,乳癌為人表皮生長因子受體2陰性(HER-)乳癌。在其他的此等實施態樣中,乳癌為人表皮生長因子受體2陽性(HER+)乳癌。在另外的實施態樣中,乳癌為可以HR+、HER2-或ER+、HER2-特徵化之晚期或轉移性乳癌。 In another aspect, the invention provides a method of treating cancer comprising administering a therapeutically effective amount of a solid dosage form of any of the conditions and embodiments described herein to a subject in need thereof. In a particular embodiment, the cancer is breast cancer. In some such embodiments, the breast cancer is a hormone receptor positive (HR+) breast cancer. In some such embodiments, the breast cancer is an estrogen receptor positive (ER+) breast cancer. In some such embodiments, the breast cancer is human epidermal growth factor receptor 2 negative (HER-) breast cancer. In other such embodiments, the breast cancer is human epidermal growth factor receptor 2 positive (HER+) breast cancer. In another embodiment, the breast cancer is advanced or metastatic breast cancer that can be characterized by HR+, HER2- or ER+, HER2-.

帕博西里可單獨或與其他的藥物組合投予,特別為芳香酶抑制劑,例如來曲唑、氟維司群或依西美坦(exemestane),且通常以與一或多種醫藥上可接受之賦形劑締合之調配物投予。術語〝賦形劑〞說明除了帕博西里或其鹽以外的任何成分。 Pabsili can be administered alone or in combination with other drugs, particularly aromatase inhibitors, such as letrozole, fulvestrant or exemestane, and are usually pharmaceutically acceptable with one or more Formulations associated with excipients are administered. The term "excipient" refers to any ingredient other than paclocil or its salt.

帕博西里可經口投予。經口投予可包含吞嚥,使得化合物進入胃腸道,或可使用頰內或舌下投予,藉此使化合物自嘴直接進入血流中。 Paphosili can be administered orally. Oral administration can include swallowing such that the compound enters the gastrointestinal tract, or can be administered intravesically or sublingually, thereby allowing the compound to enter the bloodstream directly from the mouth.

可將本發明的劑型以治療有效量投予需要此治療之受試者。如本文所使用的術語〝治療有效量〞係指欲投予之化合物量,其緩和欲治療之病症的症狀中之一或多者至某種程度。關於癌症治療的治療有效量係指具有下列效應之量:(1)縮減腫瘤尺寸,(2)抑制(亦即減慢至某種程度,較佳為停止)腫瘤轉移,(3)抑制(亦即減慢至某種程度,較佳為停止)腫瘤生長或腫瘤侵襲至某種程度,及/或(4)減緩與癌相關聯的一或多體徵或症狀至某種程度(或較佳為消除)。 The dosage form of the invention can be administered to a subject in need of such treatment in a therapeutically effective amount. The term "therapeutically effective amount" as used herein refers to an amount of a compound to be administered which relaxes one or more of the symptoms of the condition to be treated to some extent. A therapeutically effective amount for cancer treatment refers to an amount that has the following effects: (1) reducing tumor size, (2) inhibiting (i.e., slowing down to some extent, preferably stopping) tumor metastasis, and (3) inhibiting (also That is, slowing down to some extent, preferably stopping) tumor growth or tumor invasion to some extent, and/or (4) slowing down one or more signs or symptoms associated with cancer to some extent (or preferably eliminate).

如本文所使用的〝受試者〞係指人或動物受試者。在特定的較佳實施態樣中,受試者為人。在一些實施態樣中,受試者為受疾病狀態所苦之患者。在其他的實施態樣中,受試者可為健康的自願者。 A sputum sputum as used herein refers to a human or animal subject. In certain preferred embodiments, the subject is a human. In some embodiments, the subject is a patient suffering from a disease state. In other embodiments, the subject can be a healthy volunteer.

如本文所使用的術語〝治療(treating)〞意指反轉、緩解、抑制此術語適用之病症或病況或此等病症或病況的一或多個症狀的進展,或預防上述病症或病況,除非 另有其他指示。如本文所使用的術語〝治療(treatment)〞係指剛如上文所定義之治療的行為,除非另有其他指示。術語〝治療〞亦包括受試者的輔助及新輔助治療。帕博西里可單獨或與芳香酶抑制劑組合投予,諸如來曲唑、氟維司群或依西美坦。 The term "treating" as used herein means reversing, alleviating, inhibiting the progression of a condition or condition to which the term applies or the progression of one or more symptoms of such condition or condition, or preventing the above described condition or condition, unless There are other instructions. The term "treatment" as used herein refers to the act of treatment just as defined above, unless otherwise indicated. The term "treatment" also includes adjuvant and neoadjuvant treatment of the subject. Pabsili can be administered alone or in combination with an aromatase inhibitor, such as letrozole, fulvestrant or exemestane.

如本文所使用的〝癌〞係指由異常細胞所引起之任何惡性及/或侵入的生長或腫瘤。如本文所使用的〝癌〞係指以形成癌(例如乳癌)之細胞類型命名的實體腫瘤,及血液癌、骨髓癌或淋巴系統癌。實體腫瘤的實例包括但不限於肉瘤或癌。血液癌的實例包括但不限於白血病、淋巴瘤和骨髓瘤。術語〝癌〞包括但不限於源自體內的特定位置之原發性癌、自開始的地方擴散至身體其他部位的轉移性癌、自原始的原發性癌緩解後復發及第二種原發性癌(其為在與後來的癌不同類型之先前癌病史的病人之新生原發性癌)。 As used herein, sputum cancer refers to any malignant and/or invasive growth or tumor caused by abnormal cells. As used herein, a sputum cancer refers to a solid tumor named after a cell type that forms a cancer (for example, breast cancer), and a blood cancer, a bone marrow cancer, or a lymphatic system cancer. Examples of solid tumors include, but are not limited to, sarcomas or carcinomas. Examples of blood cancers include, but are not limited to, leukemia, lymphoma, and myeloma. The term "carcinoma" includes, but is not limited to, primary cancer derived from a specific location in the body, metastatic cancer that spreads from the beginning to other parts of the body, recurrence from the original primary cancer, and a second primary Sexual cancer (which is a neonatal primary cancer in a patient with a different preclinical history than a later cancer).

更特定言之,與本發明(較佳地與芳香酶抑制劑組合)有關的癌之實例尤其包括乳癌。例如,癌可為激素受體陽性(HR+)乳癌,且特別為雌激素受體陽性(ER+)乳癌。在一些實施態樣中,該ER+乳癌為人表皮生長因子2(HER2)-陰性。在另外的實施態樣中,癌為ER+、HER2-晚期轉移性乳癌,其中藥物係與治療轉移性疾病之芳香酶抑制劑組合投予。 More specifically, examples of cancers associated with the present invention, preferably in combination with an aromatase inhibitor, include, in particular, breast cancer. For example, the cancer can be a hormone receptor positive (HR+) breast cancer, and in particular an estrogen receptor positive (ER+) breast cancer. In some embodiments, the ER+ breast cancer is human epidermal growth factor 2 (HER2)-negative. In another embodiment, the cancer is ER+, HER2-advanced metastatic breast cancer, wherein the drug is administered in combination with an aromatase inhibitor for treating a metastatic disease.

適合於經口投予之調配物包括固態調配物,諸如錠劑、膠囊、粉劑、菱形錠(包括以液體填充)、囊袋及類 似者。在本發明較佳的態樣中,本文所提供的固態劑型為錠劑。在一些此等實施態樣中,錠劑經包膜。在其他的此等實施態樣中,錠劑為雙層錠劑。 Formulations suitable for oral administration include solid formulations such as lozenges, capsules, powders, diamond ingots (including filling with liquids), pouches and the like. Like. In a preferred aspect of the invention, the solid dosage form provided herein is a tablet. In some such embodiments, the tablet is encapsulated. In other such embodiments, the lozenge is a bilayer tablet.

用於錠劑劑型的帕博西里可取決於劑量而構成劑型的從1重量%至80重量%,典型地構成劑型的從5重量%至60重量%,更典型地構成劑型的從約10重量%至約35重量%,或甚至更典型地構成劑型的從約15重量%至約25重量%。在特定的實施態樣中,帕博西里構成劑型的約20重量%。 The paclitaxel used in the lozenge dosage form may comprise from 1% to 80% by weight of the dosage form depending on the dosage, typically from 5% to 60% by weight of the dosage form, more typically from about 10% by weight of the dosage form. From about 15% by weight to about 35% by weight, or even more typically from about 15% to about 25% by weight of the dosage form. In a particular embodiment, the Pabsili constitutes about 20% by weight of the dosage form.

在本發明的固態劑型中,載劑可包含各種醫藥上可接受之賦形劑,包括例如稀釋劑、崩解劑、結合劑、潤滑劑、助滑劑及表面活性劑。調配物亦可包括賦形劑,諸如保存劑、抗氧化劑、香料及著色劑,以及本技術中已知的其他賦形劑。 In the solid dosage form of the present invention, the carrier may contain various pharmaceutically acceptable excipients including, for example, diluents, disintegrating agents, binding agents, lubricants, slip agents, and surfactants. Formulations may also include excipients such as preservatives, antioxidants, perfumes, and coloring agents, as well as other excipients known in the art.

固態劑型(諸如錠劑)典型地含有稀釋劑,例如乳糖(單一水化物、噴霧乾燥之單一水化物、無水物及類似者)、甘露醇、木醣醇、葡萄糖、蔗糖、山梨醇、可壓縮糖、微晶纖維素、粉狀纖維素、澱粉、預糊化澱粉、聚葡萄糖酸鹽、聚葡萄糖、糊精、葡萄糖、麥芽糊精、鈣酸鈣、磷酸氫鈣、磷酸鈣、硫酸鈣、碳酸鎂、氧化鎂、泊洛沙姆(poloxamer)、聚環氧乙烷、羥丙基甲基纖維素及其混合物。不同類型的微晶纖維素可適用於本文所述之調配物。微晶纖維素的實例包括Avicel®類型:PH101、PH102、PH103、PH105、PH 112、PH113、PH200、 PH301,及其他類型的微晶纖維素,諸如矽化微晶纖維素(SMCC)。在一些實施態樣中,稀釋劑係選自由下列所組成之群組:微晶纖維素、乳糖單水合物、甘露醇、山梨醇、木糖醇、碳酸鎂、磷酸氫鈣、磷酸鈣或其混合物。在特定的實施態樣中,稀釋劑包含微晶纖維素。在一些實施態樣中,稀釋劑包含一或多種類型的微晶纖維素,例如Avicel® PH105、Avicel® PH200或其混合物。在一些此等實施態樣中,稀釋劑不包括乳糖單水合物。在其他的此等實施態樣中,稀釋劑包含微晶纖維素且另外包含乳糖單水合物。稀釋劑經常構成固態劑型的從約25重量%至約75重量%,且較佳地構成劑型的從約50重量%至約75重量%。 Solid dosage forms such as lozenges typically contain diluents such as lactose (single hydrate, spray dried monohydrate, anhydrate, and the like), mannitol, xylitol, glucose, sucrose, sorbitol, compressible Sugar, microcrystalline cellulose, powdered cellulose, starch, pregelatinized starch, polygluconate, polydextrose, dextrin, glucose, maltodextrin, calcium calcium phosphate, calcium hydrogen phosphate, calcium phosphate, calcium sulfate , magnesium carbonate, magnesium oxide, poloxamer, polyethylene oxide, hydroxypropyl methylcellulose, and mixtures thereof. Different types of microcrystalline cellulose are suitable for use in the formulations described herein. Examples of microcrystalline cellulose include Avicel® type: PH101, PH102, PH103, PH105, PH 112, PH113, PH200, PH301, and other types of microcrystalline cellulose, such as deuterated microcrystalline cellulose (SMCC). In some embodiments, the diluent is selected from the group consisting of microcrystalline cellulose, lactose monohydrate, mannitol, sorbitol, xylitol, magnesium carbonate, calcium hydrogen phosphate, calcium phosphate or mixture. In a particular embodiment, the diluent comprises microcrystalline cellulose. In some embodiments, the diluent comprises one or more types of microcrystalline cellulose, such as Avicel® PH105, Avicel® PH200, or mixtures thereof. In some such embodiments, the diluent does not include lactose monohydrate. In other such embodiments, the diluent comprises microcrystalline cellulose and additionally comprises lactose monohydrate. The diluent often constitutes from about 25% to about 75% by weight of the solid dosage form, and preferably from about 50% to about 75% by weight of the dosage form.

固態劑型經常含有崩解劑。崩解劑的實例包括乙醇酸澱粉鈉、羧甲基纖維素鈉、羧甲基纖維素鈣、交聯羧甲基纖維素鈉、交聯聚維酮、聚乙烯基吡咯啶酮、甲基纖維素、微晶纖維素、低碳烷基取代之羥丙基纖維素、澱粉、預糊化澱粉和藻酸鈉。在一些實施態樣中,崩解劑為交聯聚維酮。可使用任何等級的交聯聚維酮;例如CL、CL-SF和XL等級的交聯聚維酮適用於本文所述之調配物。特定的實例包括Kollidon、Kollidon CL®、Kollidon CL-M®、Polyplasdone XL®、Polyplasdone XL-10®和Polyplasdone INF-10®。在一些實施態樣中,載劑包含至少一種選自由下列所組成之群組的崩解劑:交聯聚維酮、交聯羧甲基纖維素鈉和乙醇酸澱粉鈉。在特定的實施態樣中,崩解劑為 交聯聚維酮。崩解劑經常構成劑型的從約1重量%至約25重量%,較佳為從約5重量%至約20重量%,更佳為從約5重量%至約10重量%。 Solid dosage forms often contain a disintegrant. Examples of the disintegrant include sodium starch glycolate, sodium carboxymethylcellulose, calcium carboxymethylcellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methylcellulose. , microcrystalline cellulose, low carbon alkyl substituted hydroxypropyl cellulose, starch, pregelatinized starch and sodium alginate. In some embodiments, the disintegrant is crospovidone. Any grade of crospovidone can be used; for example, CL, CL-SF, and XL grade crospovidones are suitable for use in the formulations described herein. Specific examples include Kollidon, Kollidon CL®, Kollidon CL-M®, Polyplasdone XL®, Polyplasdone XL-10®, and Polyplasdone INF-10®. In some embodiments, the carrier comprises at least one disintegrant selected from the group consisting of crospovidone, croscarmellose sodium, and sodium starch glycolate. In a particular embodiment, the disintegrant is Cross-linked povidone. The disintegrant will often comprise from about 1% to about 25% by weight of the dosage form, preferably from about 5% to about 20% by weight, more preferably from about 5% to about 10% by weight.

可使用結合劑給予錠劑調配物黏結性。適合的結合劑包括微晶纖維素、明膠、糖、聚乙二醇、天然和合成膠、聚乙烯基吡咯啶酮、預糊化澱粉、羥丙基纖維素和羥丙基甲基纖維素。在一些實施態樣中,結合劑係選自由微晶纖維素、羥丙基纖維素和羥丙基甲基纖維素所組成之群組。在特定的實施態樣中,結合劑為微晶纖維素,例如Avicel® PH105。當有結合劑的存在時,其可構成劑型的從約0重量%至約15重量%,或從約0.2重量%至約10重量%。在一些實施態樣中,結合劑構成劑型的約5重量%至約10重量%。在特別的實施態樣中,結合劑構成劑型的約10重量%。 The binding agent can be used to impart viscosity to the formulation of the tablet. Suitable binders include microcrystalline cellulose, gelatin, sugar, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinized starch, hydroxypropylcellulose, and hydroxypropylmethylcellulose. In some embodiments, the binding agent is selected from the group consisting of microcrystalline cellulose, hydroxypropyl cellulose, and hydroxypropyl methylcellulose. In a particular embodiment, the binding agent is microcrystalline cellulose, such as Avicel® PH105. When present in the presence of a binding agent, it can comprise from about 0% to about 15% by weight of the dosage form, or from about 0.2% to about 10% by weight. In some embodiments, the binding agent comprises from about 5% to about 10% by weight of the dosage form. In a particular embodiment, the binding agent comprises about 10% by weight of the dosage form.

固態劑型時常含有一或多種潤滑劑。潤滑劑的實例包括硬脂酸鎂、硬脂酸鈣、硬脂酸鋅、硬脂基富馬酸鈉、硬脂酸鎂與月桂基硫酸鈉的混合物或該等中之二或更多者的混合物。在一些實施態樣中,潤滑劑為硬脂酸鎂及/或硬脂基富馬酸鈉。在一些實施態樣中,潤滑劑為硬脂酸鎂。在一些此等實施態樣中,固態劑型為包含顆粒內及顆粒外硬脂酸鎂之錠劑。在其他的實施態樣中,固態劑型為包含顆粒內硬脂酸鎂及顆粒外硬脂基富馬酸鈉之錠劑。當有潤滑劑的存在時,其時常構成劑型的從約0.25重量%至約10重量%,較佳為從約0.5重量%至約6重量%。 Solid dosage forms often contain one or more lubricants. Examples of the lubricant include magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, a mixture of magnesium stearate and sodium lauryl sulfate or two or more of these. mixture. In some embodiments, the lubricant is magnesium stearate and/or sodium stearyl fumarate. In some embodiments, the lubricant is magnesium stearate. In some such embodiments, the solid dosage form is a lozenge comprising intragranular and extragranular magnesium stearate. In other embodiments, the solid dosage form is a lozenge comprising intragranular magnesium stearate and extragranular stearyl fumarate. When present, the lubricant will generally comprise from about 0.25 wt% to about 10 wt%, preferably from about 0.5 wt% to about 6 wt%, of the dosage form.

錠劑亦可包含助滑劑,例如二氧化矽、膠態二氧化矽、矽酸鎂、三矽酸鎂、滑石和其他形式的二氧化矽,諸如聚集矽酸鹽和水化氧化矽。在一些實施態樣中,助滑劑為二氧化矽。當有助滑劑存在時,其可構成錠劑的從約0重量%至約10重量%,較佳為從約0.2重量%至約5重量%,或從約0.5重量%至約2重量%。 Tablets may also contain slip agents such as cerium oxide, colloidal cerium oxide, magnesium ruthenate, magnesium tricaprate, talc and other forms of cerium oxide such as aggregate citrate and hydrated cerium oxide. In some embodiments, the slip agent is cerium oxide. When present in the presence of a slip agent, it may comprise from about 0% to about 10% by weight of the tablet, preferably from about 0.2% to about 5% by weight, or from about 0.5% to about 2% by weight. .

錠劑可隨意地包括表面活性劑,諸如月桂基硫酸鈉和聚山梨酸酯80。當有表面活性劑存在時,其可構成錠劑的從0重量%至10重量%,或較佳為0.2重量%至5重量%。 The tablet may optionally include a surfactant such as sodium lauryl sulfate and polysorbate 80. When present in the presence of a surfactant, it may constitute from 0% to 10% by weight of the tablet, or preferably from 0.2% to 5% by weight.

本發明的固態劑型通常根據醫藥化學中常見的方法製備。將選擇之賦形劑可與API一起併入顆粒外室或顆粒內室中任一者或二者中。 The solid dosage form of the invention is typically prepared according to methods common in pharmaceutical chemistry. The selected excipient can be incorporated into either or both of the extragranular or intragranular compartments with the API.

範例的錠劑調配物含有從約10重量%至約35重量%之帕博西里,典型為從約15重量%至約25重量%之帕博西里;從約5重量%至約15重量%之水溶性酸;從約25重量%至約75重量%之稀釋劑;從約5重量%至約10重量%之崩解劑;從約0.5重量%至約6重量%之潤滑劑;及隨意地從約0重量%至約5重量%之助滑劑;及從約0重量%至約15重量%之結合劑。 Exemplary lozenge formulations contain from about 10% to about 35% by weight of Pabbori, typically from about 15% to about 25% by weight of Pabburi; from about 5% to about 15% by weight a water-soluble acid; from about 25% by weight to about 75% by weight of a diluent; from about 5% by weight to about 10% by weight of a disintegrant; from about 0.5% by weight to about 6% by weight of a lubricant; and optionally From about 0% by weight to about 5% by weight of the slip agent; and from about 0% by weight to about 15% by weight of the binder.

另外範例的錠劑調配物含有約20重量%之帕博西里;約10重量%之水溶性酸(較佳為琥珀酸);從約50重量%至約75重量%之稀釋劑(較佳為微晶纖維素);從約5重量%至約10重量%之崩解劑(較佳為交聯聚維 酮);從約0.5重量%至約6重量%之潤滑劑(較佳為硬脂酸鎂或硬脂基富馬酸鈉或二者);隨意地從約0重量%至約5重量%之助滑劑;及隨意地從約0重量%至約15重量%結合劑。當有助滑劑存在時,其較佳為二氧化矽,且當有結合劑存在時,其較佳為成為無水結合劑的適當類型之微晶纖維素(例如Avicel® PH105)。 Further exemplary lozenge formulations contain about 20% by weight of Pabsili; about 10% by weight of water soluble acid (preferably succinic acid); from about 50% by weight to about 75% by weight of diluent (preferably Microcrystalline cellulose); from about 5% by weight to about 10% by weight of a disintegrant (preferably cross-linked poly-dimensional) Ketone); from about 0.5% by weight to about 6% by weight of a lubricant (preferably magnesium stearate or sodium stearyl fumarate or both); optionally from about 0% by weight to about 5% by weight a slip agent; and optionally from about 0% to about 15% by weight of the binder. When a slip agent is present, it is preferably cerium oxide, and when a binder is present, it is preferably a suitable type of microcrystalline cellulose (e.g., Avicel® PH105) that becomes a waterless binder.

經口投予之固態調配物可經調配成即時及/或改良釋放型。改良釋放型調配物包括延遲、持續、脈衝、控制、靶定及程式化釋放型。改良釋放型調配物的概括說明參見美國專利案號6,106,864。 Oral formulations for oral administration can be formulated for immediate and/or modified release. Modified release formulations include delayed, sustained, pulsed, controlled, targeted, and stylized release. For a general description of modified release formulations, see U.S. Patent No. 6,106,864.

呈固態形狀錠劑形式的醫藥品典型地藉由壓縮組成最終產物之材料成為所欲錠劑形式來製造。此等材料可包括活性醫藥成分以及在製造過程期間或之後賦予產物必要或有用的性質之醫藥非活性賦形劑。錠劑硬度或抗張強度可用作為錠劑成分之黏結性的量度。若錠劑不具有充分的黏結性質,則錠劑在處理時可能分裂。最終調配物可包含一或多層且可包膜或不包膜。 Pharmaceutical products in the form of solid shaped lozenges are typically made by compressing the materials that make up the final product into the desired lozenge form. Such materials may include active pharmaceutical ingredients as well as pharmaceutically inactive excipients that impart necessary or useful properties to the product during or after the manufacturing process. Tablet hardness or tensile strength can be used as a measure of the stickiness of the tablet ingredients. If the tablet does not have sufficient bonding properties, the tablet may split during processing. The final formulation may comprise one or more layers and may or may not be enveloped.

如本技術所知,粒化為用於改進調配物的處理及製造性質之方法,例如藉由增加粒徑來改進流動。粒化實質上不改變藥物的物理形式,諸如其結晶或非晶形特徵。由那些熟習本技術領域者使用各種方法來製造錠劑劑型。此等方法的實例包括乾式粒化、濕式粒化、流體床粒化和直接壓縮。所使用之方法類型可取決於下列因素而定:諸如在調配物中的活性醫藥成分之物理特徵、所使用的賦形劑類 型及最終產物的所欲物理特徵。該等方法中之各者包括涉入將劑型之成分混合的步驟。在本發明特定的實施態樣中,以乾式粒化較佳。 As is known in the art, granulation is a method for improving the handling and manufacturing properties of a formulation, such as by increasing the particle size to improve flow. Granulation does not substantially alter the physical form of the drug, such as its crystalline or amorphous character. Tablet formulations are prepared by those skilled in the art using a variety of methods. Examples of such methods include dry granulation, wet granulation, fluid bed granulation, and direct compression. The type of method used may depend on factors such as the physical characteristics of the active pharmaceutical ingredient in the formulation, the excipients used. The desired physical characteristics of the type and final product. Each of these methods includes the step of mixing the ingredients of the dosage form. In a particular embodiment of the invention, dry granulation is preferred.

混合一定量的劑型成分通常為必要的,以便具有均勻且一致的最終產物。然而,在以濕式及乾式粒化製備醫藥錠劑時,頃發現在壓縮前的成分混合程度及強度與調配物的可壓縮性及黏結性損失有關,導致錠劑硬度降低。 It is often necessary to mix a certain amount of the dosage form ingredients in order to have a uniform and consistent final product. However, when pharmaceutical tablets were prepared by wet and dry granulation, it was found that the degree of mixing and strength of the components before compression was related to the compressibility and adhesion loss of the formulation, resulting in a decrease in the hardness of the tablet.

當使用輥壓實時,可觀察到類似的結果,例如在乾式粒化方法中。可使用輥壓實作為形成於後續壓縮成錠劑的顆粒之方法。輥壓實可降低後續的劑型可壓縮性及黏結性。 Similar results were observed when rolling in real time, such as in a dry granulation process. Roll compaction can be used as a method of forming particles which are subsequently compressed into a tablet. Roll compaction reduces subsequent compressibility and adhesion of the dosage form.

乾式粒化為以壓實步驟形成顆粒之方法,接著將壓製物以分級成可容易加工的粒子。常使用此方法改進調配物的流動性質及/或密度,其可加速進一步的製造過程,諸如製錠、膠囊化及粉末填充。壓製物係自通常含有活性成分及其他賦形劑(包括潤滑劑)的粉末摻合物直接製成。 Dry granulation is a method of forming granules in a compacting step, followed by grading the compact into particles that are easily processable. This method is often used to improve the flow properties and/or density of the formulation, which can accelerate further manufacturing processes such as tableting, encapsulation, and powder filling. The compact is made directly from a powder blend which typically contains the active ingredient and other excipients, including lubricants.

使用乾式粒化技術可優於濕式粒化方法,因為較短的加工時間及成本優勢。然而,乾式粒化通常受限於其中藥物或活性成分具有適合於形成醫藥上可接受之粒化物及劑型(諸如錠劑)之物理特徵的那些情況。 The use of dry granulation technology is superior to wet granulation because of the short processing time and cost advantages. However, dry granulation is generally limited by those in which the drug or active ingredient has physical characteristics suitable for the formation of pharmaceutically acceptable granulates and dosage forms such as troches.

通常需要添加至少一種賦形劑至調配物中,且此添加促成最終產物的錠劑尺寸增加。當錠劑尺寸必須在以適合的劑型起作用的特定參數範圍內時,為了容納提高可壓實性所增加的賦形劑量而使增加的錠劑尺寸超出限度是不可 行的。因此,時常限制製造者以乾式粒化法用於每一壓縮錠劑含有低劑量活性成分之調配物,使得調配物可容納足以使乾式粒化可行的賦形劑量。 It is usually necessary to add at least one excipient to the formulation, and this addition results in an increase in the size of the lozenge of the final product. When the size of the lozenge must be within a specific range of parameters that function in a suitable dosage form, it is not possible to accommodate an increased tablet size beyond the limit in order to accommodate the increased amount of excipients that increase the compactability. OK. Thus, the manufacturer is often limited to dry granulation for each compressed tablet containing a formulation of a low dose of active ingredient such that the formulation can hold an amount of excipient sufficient to effect dry granulation.

在醫藥劑型的開發中,重要的是平衡許多不同的目標。重要的是儘可能以合乎經濟的方式製備醫藥劑型。可能希望具有包含幾個加工步驟的簡單製法。劑型亦應該使其中所含有的活性化合物對患者達到最優化的可用性。再者,劑型應該容易吞嚥。越小的劑型越容易被患者接受且可提高患者適應性。 In the development of pharmaceutical dosage forms, it is important to balance many different goals. It is important to prepare the pharmaceutical dosage form as much as possible in an economical manner. It may be desirable to have a simple process involving several processing steps. The dosage form should also provide optimal availability of the active compound contained therein to the patient. Furthermore, the dosage form should be easy to swallow. The smaller the dosage form, the easier it is to be accepted by the patient and the patient's adaptability can be improved.

將最終的醫藥組成物加工成單位劑型(例如錠劑或膠囊)且接著包裝銷售。加工步驟係取決於特別的單位劑型而改變。例如,錠劑通常在壓力下壓縮成所欲形狀及膠囊係使用簡單的填充操作。那些熟習本技術領域者非常熟悉用於製造各種單位劑型的程序。 The final pharmaceutical composition is processed into a unit dosage form (eg, a lozenge or capsule) and then packaged for sale. The processing steps vary depending on the particular unit dosage form. For example, tablets are typically compressed under pressure into the desired shape and the capsules are used in a simple filling operation. Those skilled in the art are familiar with procedures for making various unit dosage forms.

錠劑通常係藉由施加壓力至壓錠機上欲製錠之材料上而形成。調配物必須具有使材料以準確的體積進料至模槽中的良好流動性質,且具有適合於形成錠劑的可壓縮性、可壓實性及彈出性質。 Tablets are usually formed by applying pressure to the material of the tablet to be used in the tablet. The formulation must have good flow properties that allow the material to be fed into the mold cavity in an accurate volume, and have compressibility, compressibility, and pop-up properties suitable for forming a tablet.

有許多各具有不同的生產力之壓錠機,但基本功能及操作皆類似。所有的壓錠機係藉由在兩個不銹鋼衝頭(上衝頭與下衝頭)之間施加的壓力壓緊在模槽內的錠劑調配物。壓錠機通常經設計而具有盛裝且進料調配物之加料斗,進料調配物至模槽中且調整衝頭與模頭定位之進料機制,及在旋轉壓錠機中用於引導衝頭移動的凸輪軌。兩種 類型的壓錠機為單工位或單衝頭壓錠機及多工位旋轉壓錠機。有些壓錠機提供比其他更長的滯留時間,能發生使鍵結增加。其他壓錠機可提供預壓縮。 There are many presses with different productivity, but the basic functions and operations are similar. All tablet presses compress the tablet formulation in the mold cavity by the pressure applied between the two stainless steel punches (the upper punch and the lower punch). The tablet press is typically designed to have a hopper containing the feed formulation, feed the formulation into the die and adjust the feed mechanism for the punch and die positioning, and to guide the punch movement in the rotary press. Cam track. Two Types of presses are single-station or single-punch presses and multi-station rotary presses. Some presses offer longer residence times than others and can occur with increased bonding. Other presses provide pre-compression.

濕式粒化方法亦可用於製備醫藥組成物的顆粒。濕式粒化方法說明於Remington:The Science and Practice of Pharmacy,Mack Publishing Company,Easton,Pa.,19th Edition 1995中。該等及其他方法通常為那些熟習本技術領域者已知。若使用濕式粒化,則可將揮發劑在成分混合之前、期間或之後,但是在形成顆粒之前併入混合物中。例如,可將固態揮發劑在添加結合劑溶液之前、期間或之後與粉末混合物摻合。其他的固態劑型可使用包括旋轉床粒化或噴霧乾燥分散液(SDD)之技術製備。 Wet granulation methods can also be used to prepare granules of pharmaceutical compositions. Wet granulation methods are described in Remington: The Science and Practice of Pharmacy, Mack Publishing Company, Easton, Pa., 19th Edition 1995. These and other methods are generally known to those skilled in the art. If wet granulation is used, the volatizing agent can be incorporated into the mixture before, during or after mixing of the ingredients, but prior to forming the granules. For example, the solid volatile agent can be blended with the powder mixture before, during or after the addition of the binder solution. Other solid dosage forms can be prepared using techniques including rotary bed granulation or spray dried dispersion (SDD).

本發明將於下列的非限制性實施例中例證。 The invention will be exemplified in the following non-limiting examples.

實施例 Example 實施例1 Example 1 游離鹼加上琥珀酸 Free base plus succinic acid

包含與琥珀酸乾式粒化之游離鹼形式的API(帕博西里)之錠劑係使用下列程序製備。錠劑具有表1中的組成。 Tablets containing API (Pabori) in the form of the free base with dry granulation of succinic acid were prepared using the following procedure. Tablets have the composition in Table 1.

將微晶纖維素(Avicel PH105)添加至摻合機(箱型摻合機或同等物)中且在低速度下經約25轉(以12rpm經2分鐘)混合。將API添加至摻合機中,以一部分的乳糖單水合物沖洗API容器且折疊混合。將批組量的琥珀酸、乳糖單水合物、交聯聚維酮及膠態二氧化矽添加至摻合機中,其在低速度下經約180轉(以12rpm經15分鐘)混合。 Microcrystalline cellulose (Avicel PH105) was added to a blender (box blender or equivalent) and mixed at low speed for about 25 revolutions (12 minutes at 12 rpm). The API was added to the blender and the API container was rinsed with a portion of the lactose monohydrate and folded and mixed. A batch amount of succinic acid, lactose monohydrate, crospovidone and colloidal cerium oxide was added to the blender which was mixed at about 180 rpm (for 15 minutes at 12 rpm) at low speed.

將磨碎機及袋以50%之批組量的微晶纖維素(Avicel PH102)預塗佈。將來自上述之摻合物通過磨碎機。將磨碎機以剩餘部分的微晶纖維素(Avicel PH102)沖洗且將磨碎之材料自袋轉移至摻合機(箱型摻合機或同等物)中且在低速度下經約180轉(以12rpm經15分鐘)混合。顆粒內硬脂酸鎂通過適當的分級篩子篩分且添加至先前步 驟的摻合機中。將混合物在低速度下經約60轉(以12rpm經5分鐘)混合。將摻合物經輥壓實(Gerteis Minipactor或同等物),不分離或回收細料。若不使用線內磨碎,則將輥壓實之摻合物通過分別配備有1601葉輪、050G之篩目及1000或700rpm之速度的Comil U5或U10。 The attritor and bag were precoated with a 50% batch amount of microcrystalline cellulose (Avicel PH102). The blend from the above was passed through a attritor. The attritor was rinsed with the remainder of the microcrystalline cellulose (Avicel PH102) and the ground material was transferred from the bag to a blender (box blender or equivalent) and passed at about 180 rpm at low speed. (mixed at 12 rpm for 15 minutes). The intragranular magnesium stearate is sieved through a suitable grading sieve and added to the previous step In the blending machine. The mixture was mixed at low speed for about 60 revolutions (for 12 minutes at 12 rpm). The blend is compacted by roller (Gerteis Minipactor or equivalent) without separating or recovering the fines. If in-line grinding is not used, the roll compacted blend is passed through a Comil U5 or U10 equipped with a 1601 impeller, a 050G mesh, and a speed of 1000 or 700 rpm, respectively.

將顆粒外交聯聚維酮添加至先前步驟的摻合機中。將混合物在低速度下經約180轉(以12rpm經15分鐘)混合。將顆粒外硬脂酸鎂通過適當的分級篩子篩分。將其添加至先前步驟的摻合機中且經約60轉(以12rpm經5分鐘)混合。 The granules of diplomatic vidosterone were added to the blender of the previous step. The mixture was mixed at low speed for about 180 revolutions (for 12 minutes at 12 rpm). The extragranular magnesium stearate is sieved through a suitable grading sieve. It was added to the blender of the previous step and mixed over about 60 revolutions (5 minutes at 12 rpm).

使用單工位(Korsch XP 1或同等物)或旋轉壓錠機形成錠劑。 Tablets are formed using a single station (Korsch XP 1 or equivalent) or a rotary tablet press.

實施例2 Example 2 雙層錠劑調配物 Double-layer tablet formulation

製備含有活性層及酸層之雙層錠劑。活性層係由來自實施例1之粒化摻合物所組成。 A bilayer tablet containing an active layer and an acid layer is prepared. The active layer consisted of the granulated blend from Example 1.

活性層及雙層具有表2中的組成。 The active layer and the double layer have the compositions in Table 2.

酸層摻合物係藉由將微晶纖維素、酒石酸及交聯聚維酮組合至適當的分級容器中而形成。將組合物經約120轉摻合且接著通過分別配備有1601葉輪、018R之篩目及1000或700rpm之速度的Comil U5或U10。 The acid layer blend is formed by combining microcrystalline cellulose, tartaric acid, and crospovidone into a suitable grading vessel. The composition was blended at about 120 revolutions and then passed through a Comil U5 or U10 equipped with a 1601 impeller, a 018R mesh, and a 1000 or 700 rpm speed, respectively.

將磨碎之材料轉移至摻合機中且接著將組份經約120轉摻合。將硬脂酸鎂通過適當的分級篩子(1毫米篩子,US Sieve #20)篩分,接著添加至摻合機中且經約50轉摻合。 The ground material was transferred to a blender and the components were then blended over about 120 revolutions. Magnesium stearate was sieved through a suitable graded sieve (1 mm sieve, US Sieve #20), then added to the blender and blended at about 50 revolutions.

雙層錠劑係使用下列程序使用適合的旋轉雙層壓錠機(諸如Korsch XP1)形成。將活性層粒化物(根據實施例1中概述之程序形成)壓縮成625毫克標的活性層重量及經建議之6.66毫米活性層厚度。添加酸層摻合物且將酸層與活性層壓縮成985毫克所欲填充重量。 The bilayer tablet was formed using the following procedure using a suitable rotary double laminating machine such as Korsch XP1. The active layer granules (formed according to the procedure outlined in Example 1) were compressed to a weight of 625 mg of the active layer and a recommended thickness of 6.66 mm of active layer. The acid layer blend was added and the acid layer and active layer were compressed to a desired filling weight of 985 mg.

實施例3 Example 3 流體床粒化 Fluid bed granulation

流體床塗佈機(Niro MP-2 Fluid Bed Coater,配備有 MP-1球、頂部噴霧粒化安裝)使用下列條件預加熱,直到入口空氣露點(12℃)及產物碗溫度(大於或等於45℃)穩定至標的值為止。噴嘴為具有0.8毫米液體尖端的Schlick 970及噴嘴至碗底距離33公分。 Fluid bed coater (Niro MP-2 Fluid Bed Coater, equipped with The MP-1 ball, top spray granulation installation) was preheated using the following conditions until the inlet air dew point (12 ° C) and the product bowl temperature (greater than or equal to 45 ° C) stabilized to the nominal value. The nozzle was a Schlick 970 with a 0.8 mm liquid tip and a nozzle to bowl bottom distance of 33 cm.

為了形成流體床粒化物,先將琥珀酸使用研缽和杵以手動磨碎。將磨碎之粉末定期地放在#60號網篩上,使用手動搖晃,使材料通過至收集容器中。將保留在篩子上的材料送回研缽且添加更多未經磨碎之材料。持續磨碎,直到所需的量通過篩子為止。 To form a fluid bed granulate, succinic acid was first manually ground using a mortar and pestle. The ground powder was periodically placed on a #60 mesh screen and the material was passed through to the collection container using manual shaking. The material remaining on the sieve is returned to the mortar and more unground material is added. Continue to grind until the desired amount passes through the sieve.

除了琥珀酸以外,將下列的無水床組份之各者以所需的量個別通過#30號網篩選至彼等單獨的收集容器中。 In addition to succinic acid, each of the following anhydrous bed components was individually screened through the #30 mesh to their separate collection containers in the desired amounts.

結合劑懸浮液係藉由將水添加至適當的分級容器中而形成。接著將甘露醇及羥丙基纖維素添加至容器中。將溶液經最少2小時混合且看不到聚集物。 The binder suspension is formed by adding water to a suitable grading vessel. Mannitol and hydroxypropyl cellulose are then added to the container. The solution was mixed for a minimum of 2 hours and no aggregates were visible.

接著將API緩慢地添加至溶液中以形成結合劑懸浮液。將結合劑懸浮液在加工期間攪拌,直到使用為止。結合劑懸浮液含有表4中的下列組份: The API is then slowly added to the solution to form a binding agent suspension. The binder suspension is stirred during processing until use. The binder suspension contained the following components in Table 4:

在流體床產物碗溫度穩定之後,接著將無水床材料以下列順序裝填至流體床中:甘露醇、微晶纖維素、琥珀酸及交聯聚維酮。 After the fluid bed product bowl temperature has stabilized, the anhydrous bed material is then loaded into the fluid bed in the following order: mannitol, microcrystalline cellulose, succinic acid, and crospovidone.

在29-31℃之床溫度,12-30克/分鐘之噴霧速率,70-115CMH(立方公尺/小時)之氣流及1.1巴之霧化壓力下開始流體床粒化,以提供流體床粒化物。 Fluid bed granulation is initiated at a bed temperature of 29-31 ° C, a spray rate of 12-30 g/min, a flow of 70-115 CMH (m ^ 3 /hr) and an atomization pressure of 1.1 bar to provide fluid bed granulation Compound.

實施例4 Example 4 流體床粒化物錠劑 Fluid bed granule lozenge

錠劑係使用下列程序自實施例3的流體床粒化物(FBG)形成。首先藉由將顆粒通過配備有1601葉輪、018R之篩目及1500rpm之速度的Comil U5,使粒化物乾式分級。將顆粒以視覺評定儘可能均勻地進料至Comil中(2公斤粒化物以20至25分鐘)。將磨碎之顆粒通過#60號網篩且將通過篩子的材料收集在袋中及放在旁邊。將保留在#60號網篩上的材料在Comil中第二次磨碎。將磨碎之顆粒通過#60號網篩且將材料收集在袋中。將第二 次之後保留在#60號網篩上的材料使用刮勺/刮刀溫和地推壓通過篩子,直到全部通過為止。將材料添加至袋中。 Tablets were formed from the fluid bed granules (FBG) of Example 3 using the following procedure. The granulate was first dry classified by passing the granules through a Comil U5 equipped with a 1601 impeller, a 018R mesh and a speed of 1500 rpm. The particles were visually rated as homogeneously as possible into the Comil (2 kg granules for 20 to 25 minutes). The ground granules are passed through a #60 mesh screen and the material passing through the screen is collected in a bag and placed aside. The material remaining on #60 mesh was ground a second time in Comil. The ground granules were passed through a #60 mesh screen and the material was collected in a bag. Will be second The material remaining on the #60 mesh screen after the second was gently pushed through the sieve using a spatula/scraper until all passed. Add material to the bag.

在表5中給出FBG錠劑之最終錠劑摻合物: The final tablet blend of the FBG lozenge is given in Table 5:

若必要時,以來自先前步驟的磨碎之顆粒的總重量為基準計算顆粒外組份的調整重量。 If necessary, the adjusted weight of the extragranular component is calculated based on the total weight of the ground particles from the previous step.

將琥珀酸以小等分量手動磨碎(以研缽和杵)。使用足夠的量以確保足夠量的磨碎材料。將磨碎之材料通過#60號網篩且收集在新袋中。將保留在篩子上的任何材料送回具有下一等分量的研缽中。將所需量的磨碎之琥珀酸細分。 The succinic acid was manually ground in small aliquots (for mortar and pestle). Use a sufficient amount to ensure a sufficient amount of ground material. The ground material was passed through a #60 mesh screen and collected in a new bag. Any material remaining on the sieve is returned to the mortar having the next aliquot. The required amount of ground succinic acid is subdivided.

接下來,將微晶纖維素及交聯聚維酮通過#30號網篩且添加至摻合機中。將磨碎之琥珀酸及乾式分級之流體床顆粒添加至適當分級的摻合機中。裝填密度為0.39克/毫升。將混合物以16rpm(180轉)混合11.25分鐘。 Next, the microcrystalline cellulose and crospovidone were passed through a #30 mesh screen and added to the blender. The ground succinic acid and dry graded fluid bed granules are added to a suitably graded blender. The packing density was 0.39 g/ml. The mixture was mixed at 16 rpm (180 rpm) for 11.25 minutes.

接下來,將硬脂酸鎂與3至10倍(體積,以視覺評估)來自先前步驟的摻合物在適當分級的瓶中組合。將混 合物以溫和的手動搖晃約30秒而混合且接著通過30號網篩。將內容物添加至摻合機中且以16rpm(60轉)混合3.75分鐘。 Next, magnesium stearate was combined with 3 to 10 times (by volume, visually assessed) the blend from the previous step in a suitably graded bottle. Will mix The mixture was gently shaken by hand for about 30 seconds and then passed through a No. 30 mesh screen. The contents were added to the blender and mixed at 16 rpm (60 rpm) for 3.75 minutes.

將批組使用適合的壓錠機(諸如Korsch XM12)壓縮成標的規格。 The batch is compressed into the nominal specifications using a suitable tablet press, such as the Korsch XM12.

實施例5 Example 5 噴霧乾燥分散液 Spray drying dispersion

羥丙基甲基纖維素(HPMC E3 Prem)溶液之製備如下:將3.25重量%之HPMC E3溶解在90/10之甲醇/水(w/w)的溶劑摻合物中,以形成3.2重量%之HPMC溶液。將足夠量的帕博西里(API)添加至此溶液中,以形成下列組成之噴霧懸浮液:1.75重量%之API、3.25重量%之羥丙基甲基纖維素、85.5重量%之甲醇及9.5重量%之水。接著連續攪拌懸浮液,以阻止API沉降在懸浮液槽中。 The hydroxypropyl methylcellulose (HPMC E3 Prem) solution was prepared as follows: 3.25 wt% of HPMC E3 was dissolved in a 90/10 methanol/water (w/w) solvent blend to form 3.2 wt%. HPMC solution. A sufficient amount of Pabsiri (API) was added to this solution to form a spray suspension of the following composition: 1.75 wt% API, 3.25 wt% hydroxypropyl methylcellulose, 85.5% by weight methanol, and 9.5 wt. % of water. The suspension is then continuously stirred to prevent the API from settling in the suspension tank.

將噴霧乾燥器在1850克/分鐘之流速及130℃之入口乾燥氣體溫度下使用加熱之乾燥氣體(氮)預加熱。在預加熱之後噴霧90/10(w/w)之甲醇/水摻合物,直到達成穩定態熱力學條件為止。一旦噴霧乾燥器達成穩定態時,接著將噴霧懸浮液在130克/分鐘之進料速率、130℃之溶液溫度及250psi之壓力下經由急驟霧化而引入噴霧乾燥器中。在噴嘴周圍利用在60psi之壓力下的第二個氮氣流,以防止噴嘴積垢。在溫度45℃之噴霧乾燥器出口收 集粒子。 The spray dryer was preheated using a heated drying gas (nitrogen) at a flow rate of 1850 g/min and an inlet dry gas temperature of 130 °C. A 90/10 (w/w) methanol/water blend was sprayed after preheating until steady state thermodynamic conditions were reached. Once the spray dryer reached a steady state, the spray suspension was then introduced into the spray dryer via flash atomization at a feed rate of 130 grams per minute, a solution temperature of 130 ° C, and a pressure of 250 psi. A second stream of nitrogen at a pressure of 60 psi was used around the nozzle to prevent nozzle fouling. At the outlet of the spray dryer at a temperature of 45 ° C Set particles.

在收集之後,將粒子放入在40℃/15%相對濕度下操作的對流盤乾燥器中最少6小時。這使得粒子中的殘餘溶劑減少至不超過0.3重量%之殘餘甲醇。 After collection, the particles were placed in a convection tray dryer operating at 40 ° C / 15% relative humidity for a minimum of 6 hours. This reduces the residual solvent in the particles to no more than 0.3% by weight of residual methanol.

第二乾燥粒子的粒徑係以取自Malvern Instruments Ltd.of Framingham,Massachusetts的Malvern Particle Size Analyzer使用0.5至1.0巴之低分散壓力測量,其中D(4,3)為體積平均直徑;DV10為組成含有粒子之總體積的10%之直徑;DV50為組成含有粒子之總體積的50%之直徑;及DV90為組成含有粒子之總體積的90%之直徑。在表6中給出粒徑: The particle size of the second dried particles was measured using a Malvern Particle Size Analyzer from Malvern Instruments Ltd. of Framingham, Mass., using a low dispersion pressure of 0.5 to 1.0 bar, where D(4,3) is the volume average diameter; DV 10 is The composition comprises a diameter of 10% of the total volume of the particles; DV 50 is the diameter of the composition comprising 50% of the total volume of the particles; and DV 90 is the diameter of the composition comprising 90% of the total volume of the particles. The particle sizes are given in Table 6:

粒子之間距(span)(DV90-DV10)/DV50為1.96。粒子之鬆比容積為5.6毫升/克,而抽頭比容積(tapped specific volume)為3.0毫升/克。 The particle span (DV 90 - DV 10 ) / DV 50 was 1.96. The bulk ratio of the particles was 5.6 ml/g, and the tapped specific volume was 3.0 ml/g.

如以微差掃描量熱法(DSC)在低於5%相對濕度下所測量之粒子的玻璃轉換溫度(Tg)為117.5℃。粉末X射線繞射(PXRD)顯示API為非晶形,沒有可檢測的結晶度。如以掃描電子顯微法(SEM)所測量之粒子形態顯 示具有完整及塌陷球體之粒子。 The glass transition temperature (Tg) of the particles as measured by differential scanning calorimetry (DSC) at less than 5% relative humidity was 117.5 °C. Powder X-ray diffraction (PXRD) shows that the API is amorphous with no detectable crystallinity. Such as particle morphology measured by scanning electron microscopy (SEM) Show particles with intact and collapsed spheres.

實施例6 Example 6 比較用噴霧乾燥分散液錠劑 Comparative spray-dried dispersion lozenge

缺少水溶性酸之錠劑係使用下列程序自實施例5之噴霧乾燥分散液(SDD)形成。將半量的微晶纖維素添加至摻合機(箱型摻合機或同等物)中且在低速度下經約25轉(以12rpm經2分鐘)混合。將SDD添加至摻合機中。將摻合機以一部分的氯化鈉沖洗且混合。 Tablets lacking a water soluble acid were formed from the spray dried dispersion (SDD) of Example 5 using the following procedure. A half amount of microcrystalline cellulose was added to the blender (box blender or equivalent) and mixed at low speed for about 25 revolutions (2 minutes at 12 rpm). Add SDD to the blender. The blender was rinsed with a portion of sodium chloride and mixed.

添加批組量的氯化鈉、交聯羧甲基纖維素鈉及膠態二氧化矽且在低速度下經約180轉(以12rpm經15分鐘)混合。在表7中給出最終摻合物: A batch amount of sodium chloride, croscarmellose sodium, and colloidal ceria was added and mixed at about 180 rpm (for 15 minutes at 12 rpm) at low speed. The final blend is given in Table 7:

將磨碎機及袋以25重量%之量的微晶纖維素預塗 佈。將來自上述步驟之摻合物通過使用配備有1601葉輪、032R之篩目及1000之速度的Comil U5之磨碎機。將磨碎機以剩餘批組部分的微晶纖維素沖洗且將磨碎之材料自袋轉移至摻合機(箱型摻合機或同等物)中且在低速度下經約180轉(以12rpm經15分鐘)混合。 Pre-coating the attritor and bag with microcrystalline cellulose in an amount of 25% by weight cloth. The blend from the above procedure was passed through a grinder equipped with a 1601 impeller, a mesh of 032R and a Comil U5 at a speed of 1000. The attritor is rinsed with the remaining batch portion of microcrystalline cellulose and the ground material is transferred from the bag to a blender (box blender or equivalent) and at about 180 revolutions at low speed ( Mix at 12 rpm for 15 minutes).

顆粒內硬脂酸鎂通過適當的分級篩子篩分且添加至先前步驟的摻合機中,接著在低速度下經約60轉(以12rpm經5分鐘)混合。 The intragranular magnesium stearate was sieved through a suitable grading sieve and added to the blender of the previous step, followed by mixing at about 60 revolutions (5 minutes at 12 rpm) at low speed.

將摻合物使用Korsch XP 1或同等物壓實。 The blend was compacted using Korsch XP 1 or equivalent.

將壓實之摻合物通過使用配備有1601葉輪、050G之篩目及1000之速度的Comil U5之磨碎機。 The compacted blend was passed through a grinder equipped with a 1601 impeller, a 050 G mesh and a speed of 1000 Comil U5.

將粒化物自袋轉移至摻合機(箱型摻合機或同等物)中。計算顆粒外膠態二氧化矽的量且添加至先前步驟的摻合機中,接著經約180轉(以12rpm經15分鐘)混合。計算所需之顆粒外硬脂酸鎂量,通過適當的分級篩子篩分且添加至先前步驟的摻合機中,接著經約60轉(以12rpm經5分鐘)混合。 The granulate is transferred from the bag to a blender (box blender or equivalent). The amount of extragranular colloidal ceria was calculated and added to the blender of the previous step, followed by mixing at about 180 revolutions (15 minutes at 12 rpm). The amount of extragranular magnesium stearate required was calculated, sieved through a suitable graded sieve and added to the blender of the previous step, followed by mixing at about 60 revolutions (5 minutes at 12 rpm).

將錠劑使用單工位壓縮機(Korsch XP 1或同等物)壓實。 The tablets were compacted using a single station compressor (Korsch XP 1 or equivalent).

實施例7 Example 7 噴霧乾燥分散液錠劑 Spray dried dispersion lozenge

錠劑係使用下列程序自實施例5之SDD形成。將半量的微晶纖維素添加至摻合機(箱型摻合機或同等物)中 且在低速度下經約25轉(以12rpm經2分鐘)混合。將SDD添加至摻合機中,將摻合機以一部分的氯化鈉沖洗且混合。添加批組量的琥珀酸、氯化鈉、交聯羧甲基纖維素鈉及膠態二氧化矽且在低速度下經約180轉(以12rpm經15分鐘)混合。在表8中給出摻合物的最終組成。 Tablets were formed from the SDD of Example 5 using the following procedure. Add half the amount of microcrystalline cellulose to the blender (box blender or equivalent) And mixing at low speed for about 25 revolutions (2 minutes at 12 rpm). The SDD was added to the blender and the blender was rinsed and mixed with a portion of sodium chloride. A batch amount of succinic acid, sodium chloride, croscarmellose sodium, and colloidal cerium oxide was added and mixed at about 180 rpm (for 15 minutes at 12 rpm) at low speed. The final composition of the blend is given in Table 8.

將磨碎機及袋以25重量%之量的微晶纖維素預塗佈。將來自上述步驟之摻合物通過使用配備有1601葉輪、018R之篩目及1000之速度的Comil U5之磨碎機。將磨碎機以剩餘批組部分的微晶纖維素沖洗,將磨碎之材料自袋轉移至摻合機(箱型摻合機或同等物)中且在低速度下經約180轉(以12rpm經15分鐘)混合。 The attritor and bag were precoated with microcrystalline cellulose in an amount of 25% by weight. The blend from the above procedure was passed through a grinder equipped with a 1601 impeller, a 018R mesh, and a 1000 speed Comil U5. The attritor is rinsed with the remaining batch portion of microcrystalline cellulose, and the ground material is transferred from the bag to a blender (box blender or equivalent) and at about 180 revolutions at low speed ( Mix at 12 rpm for 15 minutes).

顆粒內硬脂酸鎂通過適當的分級篩子篩分且添加至先 前步驟的摻合機中,接著在低速度下經約60轉(以12rpm經5分鐘)混合。 The intragranular magnesium stearate is sieved through a suitable grading sieve and added first The blender of the previous step was then mixed at about 60 revolutions (5 minutes at 12 rpm) at low speed.

將摻合物使用Korsch XP 1或同等物壓實。 The blend was compacted using Korsch XP 1 or equivalent.

將壓實之摻合物通過使用配備有1601葉輪、050G之篩目及1000之速度的Comil U5之磨碎機。計算所需之顆粒外硬脂酸鎂量,通過適當的分級篩子篩分且添加至先前步驟的摻合機中。接著將混合物經約60轉(以12rpm經5分鐘)混合。 The compacted blend was passed through a grinder equipped with a 1601 impeller, a 050 G mesh and a speed of 1000 Comil U5. The amount of extragranular magnesium stearate required is calculated, sieved through a suitable grading sieve and added to the blender of the previous step. The mixture was then mixed over about 60 revolutions (for 12 minutes at 12 rpm).

將錠劑使用單工位壓縮機(Korsch XP 1或同等物)壓實。 The tablets were compacted using a single station compressor (Korsch XP 1 or equivalent).

實施例8 Example 8 在pH 5.5下的溶解試驗1 Dissolution test at pH 5.5

製備用於溶解測試之試驗錠劑調配物,其包含游離鹼形式之帕博西里(API)、水溶性酸、微晶纖維素(Avicel PH102)、乳糖單水合物(Fast Flo 316)、交聯聚維酮(Kollidon CL)及硬脂酸鎂。使用實施例1中所述之乾式粒化(DG)方法以下列的酸製備試驗錠劑:蘋果酸、順丁烯二酸、琥珀酸、反丁烯二酸、酒石酸、甲苯磺酸、苯甲酸及苯磺酸。經由直接壓縮(DC)製備(不以乾式粒化)蘋果酸及檸檬酸之試驗錠劑。 Preparation of a test lozenge formulation for dissolution testing comprising free base form of Pabsiri (API), water soluble acid, microcrystalline cellulose (Avicel PH 102), lactose monohydrate (Fast Flo 316), cross-linking Povidone (Kollidon CL) and magnesium stearate. The test tablet was prepared using the dry granulation (DG) method described in Example 1 with the following acids: malic acid, maleic acid, succinic acid, fumaric acid, tartaric acid, toluenesulfonic acid, benzoic acid And benzenesulfonic acid. A test lozenge of malic acid and citric acid was prepared via direct compression (DC) (not dry granulation).

錠劑係在具有於50rpm自旋之葉片的USP 2旋轉裝置中於500毫升10mM pH 5.5之乙酸鈉緩衝液中及在37℃之溫度下進行溶解測試。在各拉點收集6毫升樣品且通 過10微米全流式過濾器。在367奈米之UV波長下進行離線分析。 The tablets were subjected to a dissolution test in a USP 2 rotating apparatus having blades of spin at 50 rpm in 500 ml of 10 mM sodium acetate buffer pH 5.5 and at a temperature of 37 °C. Collect 6 ml samples at each pull point and pass Over 10 micron full flow filter. Off-line analysis was performed at a UV wavelength of 367 nm.

比較用溶解數據係使用實施例1之乾式粒化方法使用缺少水溶性酸之摻合物所製備的帕博西里羥乙磺酸鹽(ISE)膠囊及游離鹼API錠劑而產生。將包含琥珀酸、順丁烯二酸、蘋果酸、反丁烯二酸及酒石酸之錠劑的溶解試驗之結果顯示於圖1中。包含琥珀酸、蘋果酸和酒石酸之錠劑展現優越的溶解性能,在30分鐘具有大於50%之藥物溶解,如圖2中所示。 The comparative dissolution data was generated using the dry granulation method of Example 1 using a Pabsiodonium isethionate (ISE) capsule prepared from a blend lacking a water-soluble acid and a free base API tablet. The results of a dissolution test of a tablet containing succinic acid, maleic acid, malic acid, fumaric acid, and tartaric acid are shown in Fig. 1. Tablets containing succinic acid, malic acid, and tartaric acid exhibited superior solubility properties with greater than 50% drug dissolution at 30 minutes, as shown in FIG.

實施例9 Example 9 調配物之化學穩定性 Chemical stability of the formulation

將實施例8中所製備之試驗錠劑在70℃/75% RH下貯存8天。將錠劑壓碎且使用如下的高性能液相層析術(HLPC)方法分析雜質:Waters CSH C18,2.1 x 100毫米,1.7微米管柱;移動相(梯度溶析液)A:0.03%三氟乙酸,及B:在乙腈中的0.03%三氟乙酸;45℃之管柱溫度;0.5毫升/分鐘之流速;在234奈米下的UV檢測;2微升注射體積;及10.72分鐘之操作時間。將結果總結於表9中。 The test tablet prepared in Example 8 was stored at 70 ° C / 75% RH for 8 days. The tablet was crushed and analyzed for impurities using the following high performance liquid chromatography (HLPC) method: Waters CSH C18, 2.1 x 100 mm, 1.7 micron column; mobile phase (gradient elution) A: 0.03% three Fluoroacetic acid, and B: 0.03% trifluoroacetic acid in acetonitrile; column temperature at 45 ° C; flow rate of 0.5 ml / min; UV detection at 234 nm; injection volume of 2 μl; and operation of 10.72 minutes time. The results are summarized in Table 9.

包含蘋果酸、琥珀酸、苯甲酸、甲苯磺酸、酒石酸及苯磺酸之錠劑在70℃/75% RH下貯存8天之後具有可接受的總雜質。以優越的溶解及穩定性實驗為基準選擇包含琥珀酸、蘋果酸和酒石酸之調配物進一步發展。 Tablets containing malic acid, succinic acid, benzoic acid, toluenesulfonic acid, tartaric acid, and benzenesulfonic acid have acceptable total impurities after storage for 8 days at 70 ° C / 75% RH. Formulations containing succinic acid, malic acid, and tartaric acid were further developed based on superior dissolution and stability experiments.

實施例10 Example 10 非漏槽(non-sink)溶解試驗2 Non-sink dissolution test 2

發展用於篩選調配物之非漏槽試管內溶解方法。在此方法中,將錠劑放入以50rpm攪拌之USP 2(葉片)裝置及在37℃之溫度下500毫升50mM磷酸鹽緩衝液+0.1M NaCl(pH 6.5)中。定期收集樣品且經由10微米過濾器過濾。在367奈米之UV下離線測量API濃度。以實施例1(FB+琥珀酸,錠劑)、實施例2(FB+琥珀酸,雙層錠劑)、實施例4(FB+琥珀酸,FBG錠劑)、實施例6(SDD,錠劑)及實施例7(SDD+琥珀 酸,錠劑)所製備之錠劑產生溶劑數據。溶解輪廓顯示於圖3中。亦顯示具有表10之調配物的市售帕博西里游離鹼膠囊(游離鹼膠囊)的溶解數據: Development of a non-leaky in-tube dissolution method for screening formulations. In this method, the tablet was placed in a USP 2 (blade) device stirred at 50 rpm and in a solution of 500 ml of 50 mM phosphate buffer + 0.1 M NaCl (pH 6.5) at a temperature of 37 °C. Samples were collected periodically and filtered through a 10 micron filter. The API concentration was measured offline under UV of 367 nm. Example 1 (FB+ succinic acid, lozenge), Example 2 (FB+ succinic acid, bilayer tablet), Example 4 (FB+ succinic acid, FBG lozenge), Example 6 (SDD, lozenge) and The tablets prepared in Example 7 (SDD + succinic acid, lozenges) produced solvent data. The dissolution profile is shown in Figure 3. Dissolution data for commercially available Pabsiri free base capsules (free base capsules) with the formulation of Table 10 are also shown:

如圖3中所市,施予非漏槽溶解條件的實施例1、2和4之劑型溶解(a)在15分鐘內不少於40%之帕博西里;(b)在30分鐘內不少於35%之帕博西里;(c)在60分鐘內不少於25%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 As shown in Fig. 3, the dosage forms of Examples 1, 2 and 4 which are subjected to the non-leakage dissolution conditions are (a) not less than 40% of Pabori in 15 minutes; (b) not within 30 minutes. Less than 35% of Pabsiri; (c) not less than 25% of Pabori in 60 minutes; or (d) two or more of (a), (b) and (c).

實施例11 Example 11 市售游離鹼膠囊之藥物暴露程度 Drug exposure of commercially available free base capsules

隨機、單劑量、開放標籤、4順序、4期交叉研究係在健康的志願者中操作。二十八(28)位受試者在4個不同條件或治療下(隔夜空腹[A]、給藥前高脂肪膳食[B]、給藥前低脂肪膳食[C]及在給藥前1小時與給藥後2小時 中脂肪膳食[D])各接受125毫克單一劑量帕博西里。 Random, single-dose, open-label, 4-sequence, and 4-phase crossover studies were performed in healthy volunteers. Twenty-eight (28) subjects under 4 different conditions or treatments (overnight fast [A], pre-dose high-fat diet [B], pre-dose low-fat diet [C] and before dosing 1 Hours and 2 hours after dosing The medium fat diet [D]) each received a single dose of 125 mg of Pabsiri.

將第1期至第4期所使用之治療順序呈示於表11中。在研究期之間有至少10天的洗除期。在治療投予之後,使受試者進行144小時之PK取樣。在表12中給出藥物動力學參數值。 The treatment sequences used in the first to fourth phases are shown in Table 11. There is at least a 10 day washout period between study periods. Subjects were allowed to undergo 144 hours of PK sampling after treatment administration. The pharmacokinetic parameter values are given in Table 12.

以125毫克帕博西里市售膠囊調配物在空腹條件下投予時展現比三個進食條件下的暴露程度低的AUC及Cmax。因此,建議市售膠囊與食物一起投予。 When the 125 mg Pabsiri commercially available capsule formulation was administered under fasting conditions, it exhibited lower AUC and Cmax than the exposure under three eating conditions. Therefore, it is recommended that commercially available capsules be administered together with food.

實施例12 Example 12 制酸劑對帕博西里之生物利用率的效應 Effect of antacids on bioavailability of pabexili

此研究的目的係調查以多重劑量的質子泵抑制劑(PPI,尤其為瑞貝拉唑)治療所達成的胃pH增加對在空腹條件下給予125毫克單一經口帕博西里市售膠囊之藥物動力學(PK)的可能效應。 The aim of this study was to investigate the increase in gastric pH achieved by treatment with multiple doses of proton pump inhibitors (PPI, especially rabeprazole) for the administration of 125 mg of a single oral paclicil capsule under fasting conditions. Possible effects of kinetics (PK).

將結果總結於以下表13中。125毫克帕博西里市售膠囊調配物在瑞貝拉唑的存在下投予時展現比單獨投予的帕博西里顯著更低的AUC及Cmax。 The results are summarized in Table 13 below. The 125 mg Pabsiri commercially available capsule formulation exhibited significantly lower AUC and Cmax than the separately administered paabutir when administered in the presence of rabeprazole.

實施例13 Example 13 制酸劑對試驗調配物之生物利用率的效應 Effect of antacids on bioavailability of test formulations

在健康的志願者中進行交叉、開放標籤、非隨機藥物動力學研究以評估在空腹條件下制酸劑處理對帕博西里的六個實驗調配物之125毫克錠劑(單一劑量)在瑞貝拉唑的存在下相對於單獨投予的帕博西里之生物利用率的效 應。表14(A)-(F)分別顯示1-6組:實施例1、實施例6、實施例7、實施例2、實施例4及125毫克帕博西里經口溶液的結果。 Cross-over, open-label, non-random pharmacokinetic studies were performed in healthy volunteers to evaluate 125 mg of lozenges (single dose) of six experimental formulations of paclitaxel treated with antacids under fasting conditions in Rebecca Effect of the bioavailability of Pabsiri administered separately in the presence of latazole should. Tables 14(A)-(F) show the results of the 1-6 groups: Example 1, Example 6, Example 7, Example 2, Example 4, and 125 mg of Pabsiri oral solution.

實施例14 Example 14 加速之穩定性試驗 Accelerated stability test

將實施例1之錠劑(無水粒化物+琥珀酸)使用下列條件加諸加速穩定性。將錠劑放入在以下列示之條件下的燒杯中。濕度控制係通過烘箱控制或飽和鹽溶液而達成。定期自下列表15中所示之條件移出樣品。將樣品(包括未暴露之對照物)貯存在冰箱中,直到分析為止。 The tablet of Example 1 (anhydrous granule + succinic acid) was subjected to accelerated stability using the following conditions. The tablets were placed in a beaker under the conditions shown below. Humidity control is achieved by oven control or saturated salt solution. Samples were removed periodically from the conditions shown in Table 15 below. Samples (including unexposed controls) were stored in the refrigerator until analysis.

以下列的飽和鹽維持指定的相對濕度:經由乙酸鉀控制22% RH;經由溴化鈉控制在30℃及40℃條件下的50% RH;及經由濕度控制烘箱控制75% RH。 The specified relative humidity was maintained with the following saturated salts: 22% RH via potassium acetate; 50% RH at 30 °C and 40 °C via sodium bromide; and 75% RH via a humidity controlled oven.

降解物為具有以下結構的API之琥珀醯基加成物: The degradant is an amber oxime-based adduct of API having the following structure:

樣品的製備如下:將各錠劑添加至100毫升量瓶中,添加攪拌棒及100毫升溶解溶劑(1440毫升水:400毫升乙腈:160毫升1N HCl;80:20之0.1N HCl:乙腈)。將燒瓶放在攪拌盤上且將樣品以最高的可用攪拌速度攪拌1小時。接著移出等分量且在聚丙烯管中以3000RPM離心5分鐘。將上清液經溶解溶劑以1:5稀釋且取樣至用於分析的HPLC小瓶中。0.25毫克/毫升之最終溶液濃度的各錠劑含125毫克API。樣品係使用下列HPLC方法分析。將結果呈示於以下表15中且顯示極佳的長期穩定性。 The samples were prepared as follows: Each tablet was added to a 100 ml volumetric flask, and a stir bar and 100 ml of dissolved solvent (1440 ml of water: 400 ml of acetonitrile: 160 ml of 1 N HCl; 80:20 of 0.1 N HCl: acetonitrile) were added. The flask was placed on a stir plate and the sample was stirred for 1 hour at the highest available agitation speed. The aliquot was then removed and centrifuged at 3000 RPM for 5 minutes in a polypropylene tube. The supernatant was diluted 1 :5 with dissolved solvent and sampled into an HPLC vial for analysis. Each tablet at a final solution concentration of 0.25 mg/ml contained 125 mg of API. The samples were analyzed using the following HPLC methods. The results are presented in Table 15 below and show excellent long term stability.

溶解溶劑(稀釋劑):1440毫升水:400毫升乙腈:160毫升1N HCl///(80:20之0.1N HCl:乙腈) Dissolving solvent (diluent): 1440 ml water: 400 ml acetonitrile: 160 ml 1 N HCl / / / (80: 20 0.1 N HCl: acetonitrile)

移動相: Mobile phase:

移動相A:0.03%三氟乙酸(在2000毫升水中的0.6毫升三氟乙酸) Mobile phase A: 0.03% trifluoroacetic acid (0.6 ml trifluoroacetic acid in 2000 ml water)

移動相B:在乙腈中的0.03%三氟乙酸(在2000毫升 乙腈中的0.6毫升三氟乙酸) 0.5毫升/分鐘 Mobile phase B: 0.03% trifluoroacetic acid in acetonitrile (in 2000 ml) 0.6 ml of trifluoroacetic acid in acetonitrile) 0.5 ml / min

梯度 gradient

234奈米,面積%相對帕博西里(主峰)2微升樣品注射液(如下文指示方式製備)。樣品室維持在5℃ 234 nm, area % relative to Pabsiri (main peak) 2 μl sample injection (prepared as indicated below). The sample chamber is maintained at 5 ° C

樣品製備 Sample Preparation

樣品的製備如下:將各錠劑添加至100毫升量瓶中,添加攪拌棒及100毫升溶解溶劑。將燒瓶放在攪拌盤上且將樣品以最高的可用攪拌速度攪拌1小時。接著移出等分量且在聚丙烯管中以3000RPM離心5分鐘。將上清液經溶解溶劑以1:5稀釋且取樣至用於分析的HPLC小瓶中。0.25毫克/毫升之最終溶液濃度的各錠劑含有125毫克API。 The samples were prepared as follows: Each tablet was added to a 100 ml volumetric flask, and a stir bar and 100 ml of dissolution solvent were added. The flask was placed on a stir plate and the sample was stirred for 1 hour at the highest available agitation speed. The aliquot was then removed and centrifuged at 3000 RPM for 5 minutes in a polypropylene tube. The supernatant was diluted 1 :5 with dissolved solvent and sampled into an HPLC vial for analysis. Each tablet at a final solution concentration of 0.25 mg/ml contained 125 mg of API.

另外,實施例4的錠劑(流體床粒化物+琥珀酸)係以相同的方式分析。將結果呈示於以下表16中且顯示對長期穩定性不可接受的降解物濃度。 Further, the tablet (fluid bed granule + succinic acid) of Example 4 was analyzed in the same manner. The results are presented in Table 16 below and show the concentration of degradants that are unacceptable for long term stability.

實施例15 Example 15 長期貯存穩定性 Long-term storage stability

將實施例1的錠劑與乾燥劑盒使用熱感應密封包裝在瓶內。在25℃/60% RH下貯存1年之後,錠劑具有少於0.05%之琥珀醯基加成物量。將實施例4的錠劑與乾燥劑盒使用熱感應密封包裝在HDPE瓶內。在25℃/60% RH下貯存6個月之後,錠劑具有少於0.23%之琥珀醯基加成物量。 The tablet and desiccant cartridge of Example 1 were packaged in a bottle using a heat sensitive seal. After storage for 1 year at 25 ° C / 60% RH, the tablet has less than 0.05% amber thiol addition. The tablet and desiccant cartridge of Example 4 were packaged in a HDPE bottle using a heat sensitive seal. After 6 months storage at 25 ° C / 60% RH, the tablet has an amount of amber oxime addition of less than 0.23%.

實施例16 Example 16 包膜錠劑調配物 Coated lozenge formulation

表17說明最優化調配物A1、A2及B,其顯示固態劑型商業化可接受之製造性能。錠劑係使用下列程序形成。將微晶纖維素PH102、膠態二氧化矽、顆粒內交聯聚維酮CL及API摻合在一起且通過Comil而均勻化。將顆粒內硬脂酸鎂摻合至其中。使用輥壓實及顆粒磨碎使混合物經乾式粒化。 Table 17 illustrates the optimization of formulations A1, A2, and B, which show that the solid dosage form is commercially acceptable for manufacturing performance. Tablets were formed using the following procedure. Microcrystalline cellulose PH102, colloidal cerium oxide, intragranular cross-linked povidone CL and API were blended together and homogenized by Comil. The intragranular magnesium stearate is blended therein. The mixture was dry granulated using roller compaction and particle milling.

將篩分之琥珀酸、微晶纖維素PH-200及顆粒外交聯聚維酮(CL或CL-SF級)摻合至顆粒中。將作為製錠潤滑劑的顆粒外硬脂酸鎂或硬脂基富馬酸鈉混合至最終摻合物中。使用具有預壓縮的旋轉壓錠機形成錠劑。調配物A2使用外部潤滑系統(ELS),以標稱量潤滑劑直接施加於錠劑衝頭。 The sieved succinic acid, microcrystalline cellulose PH-200, and granule diplomatic ketoxime (CL or CL-SF grade) are blended into the granules. The extragranular magnesium stearate or sodium stearyl fumarate as a tableting lubricant is mixed into the final blend. A tablet is formed using a rotary tablet press having a pre-compression. Formulation A2 uses an external lubrication system (ELS) to apply the lubricant directly to the tablet punch at a nominal amount.

將錠劑使用固態含量的12% w/w之Opadry Pink(03K140024)及純化水包膜至2至4%增重。 The tablets were coated with a solids content of 12% w/w Opadry Pink (03K140024) and purified water to a 2 to 4% weight gain.

實施例17 Example 17 最優化潤滑劑含量 Optimized lubricant content

調配物係如實施例16中用於調配物A2所述方式以不同量的顆粒內及顆粒外硬脂酸鎂潤滑劑製備,如表17中所示。測量各調配物的硬度(錠劑破碎力,USP<1217>)、易脆性(USP<1216>經100轉)、延展易脆性(USP<1216>經375轉)及衝頭黏附性(Hutchins,MacDonald,Mullarney,Assessing Tablet-Sticking Propensity,Pharmaceutical Technology,Volume 36,Issue 1,2012),如表18中所記述。以縮減的樣品尺寸用於USP方法。 Formulations were prepared in different amounts of intragranular and extragranular magnesium stearate lubricant as described for use in Formulation A2 in Example 16, as shown in Table 17. The hardness of each formulation (slurry breaking force, USP<1217>), brittleness (USP<1216>100 rpm), stretch brittleness (USP<1216>375 rpm) and punch adhesion were measured (Hutchins, MacDonald, Mullarney, Assessing Tablet-Sticking Propensity, Pharmaceutical Technology, Volume 36, Issue 1, 2012), as described in Table 18. The reduced sample size was used in the USP method.

減少顆粒內及顆粒外潤滑劑含量顯著地增加錠劑硬度及降低易脆性。錠劑黏附性對顆粒外潤滑劑含量比對顆粒內潤滑劑含量更敏感。以增加顆粒外硬脂酸鎂含量使黏附性顯著降低。以不同的顆粒內硬脂酸鎂含量未觀察到差別的滾紋/平滑輥黏附性。如表18中所示,從1重量%/0重量%減少至0.33重量%/0.5重量%的顆粒內/顆粒外潤滑劑配給量供給良好的錠劑強度、降低的易脆性及降低對調配物A2之錠劑的黏附性。約5重量%之硬脂基富馬酸鈉或2重量%之細粒級硬脂酸鹽(CaSt 2249)的潤滑劑含量減少衝頭黏附性及錠劑缺陷。 Reducing the amount of intragranular and extragranular lubricant significantly increases tablet hardness and reduces brittleness. Lozenge adhesion is more sensitive to the amount of extragranular lubricant than to the amount of lubricant in the granule. The adhesion is significantly reduced by increasing the extragranular magnesium stearate content. No difference in knurling/smoothing roll adhesion was observed with different intragranular magnesium stearate content. As shown in Table 18, the amount of intragranular/extragranular lubricant dispensed from 1% by weight/0% by weight to 0.33% by weight/0.5% by weight provides good tablet strength, reduced brittleness and reduced formulation. Adhesion of the A2 tablet. A lubricant content of about 5% by weight of sodium stearyl fumarate or 2% by weight of fine-grained stearate (CaSt 2249) reduces punch adhesion and tablet defects.

實施例18 Example 18 最優化錠劑硬度及易脆性 Optimized tablet hardness and brittleness

調配物係如實施例16中用於調配物12之概述方式製備,改變添加酸、添加乳糖單水合物、添加無水結合劑(Kollidon SF,Avicel PH105,Klucel EXF)的順序及改變顆粒內微晶纖維素對顆粒外微晶纖維素之比。測量該等不同的調配物之性質:最初摻合物裝填密度(USP<616>)、經輥壓實之帶狀物抗張強度(Zinchuk AV,Mullarney MP,Hancock BC.Simulation of roller compaction using a laboratory scale compaction simulator.Int J Pharm.2004 Jan 28;269(2):403-15.)、衝頭黏附性(Hutchins,MacDonald,Mullarney,Assessing Tablet-Sticking Propensity,Pharmaceutical Technology,Volume 36,Issue 1,2012)、錠劑硬度(錠劑破碎力,USP<1217>)、易脆性(USP<1216>經375轉)及崩解時間 (USP<701>)。以縮減的樣品尺寸用於USP方法。 Formulations were prepared as outlined in Example 16 for Formulation 12, changing the order of addition of acid, addition of lactose monohydrate, addition of anhydrous binder (Kollidon SF, Avicel PH105, Klucel EXF) and modification of intragranular crystallites. The ratio of cellulose to extragranular microcrystalline cellulose. The properties of the different formulations were measured: initial blend loading density (USP <616>), roll compacted tensile strength (Zinchuk AV, Mullarney MP, Hancock BC. Simulation of roller compaction using a Laboratory scale compaction simulator. Int J Pharm. 2004 Jan 28; 269(2): 403-15.), punch adhesion (Hutchins, MacDonald, Mullarney, Assessing Tablet-Sticking Propensity, Pharmaceutical Technology, Volume 36, Issue 1, 2012), tablet hardness (slurry breaking force, USP<1217>), fragility (USP<1216> via 375 rpm) and disintegration time (USP<701>). The reduced sample size was used in the USP method.

沒有乳糖的調配物顯示更高的錠劑硬度及更低的易脆性。另外,添加SF(超細粒)級Kollidon有助於降低崩解時間,同時增加錠劑硬度。經發現於顆粒外添加酸為獲得較短的崩解時間之優選。 Formulations without lactose showed higher tablet hardness and lower brittleness. In addition, the addition of SF (ultrafine particle) grade Kollidon helps to reduce the disintegration time while increasing the tablet hardness. It has been found that the addition of an acid outside the particles is preferred to achieve a shorter disintegration time.

自調配物移除乳酸提供具有降低易脆性的錠劑,其仍維持快速崩解。內含無水結合劑不提供降低易脆性的額外好處,但是些微地降低黏附性。然而,內含結合劑可在一些情況中對崩解/膨脹有負面衝擊。 Self-formulation removal of lactic acid provides a lozenge with reduced fragility that still maintains rapid disintegration. The inclusion of a water-free binder does not provide the added benefit of reducing brittleness, but slightly reduces adhesion. However, the inclusion of a binder may have a negative impact on disintegration/expansion in some cases.

實施例19 Example 19 調配物A1、A2及B之試管內溶解 In-vitro dissolution of formulations A1, A2 and B

最優化調配物A1、A2及B之試管內溶解係使用溶解試驗2在實施例10中所述之非漏槽狀態(pH 6.5,50mM磷酸鹽緩衝液,0.1M NaCl)下測定。將該等調配物之溶解輪廓與根據實施例1至7中所製備的錠劑比較。實施例1的錠劑顯示在人類志願者的空腹狀態下與質子泵抑制劑沒有藥物-藥物交互作用且設定為最小的標的溶解輪廓。將比較的溶解數據呈示於圖4中。調配物A1、A2及B全部皆優於市售游離鹼膠囊(如實施例10中所述而製得)及在早期發展中所使用之具有表20中的調配物之帕博西里羥乙磺酸鹽(ISE)膠囊二者。在10分鐘之後,調配物A1、A2及B全部皆優於實施例1至7的錠劑,且因此符合固態劑型溶解標的。 The in-tube dissolution of the optimized formulations A1, A2 and B was determined using Dissolution Test 2 in the non-leaky state (pH 6.5, 50 mM phosphate buffer, 0.1 M NaCl) described in Example 10. The dissolution profile of the formulations was compared to the tablets prepared according to Examples 1 to 7. The lozenge of Example 1 showed no drug-drug interaction with the proton pump inhibitor in the fasting state of the human volunteer and was set to the minimum target dissolution profile. The comparative dissolution data is presented in Figure 4. Formulations A1, A2, and B were all superior to commercially available free base capsules (as described in Example 10) and were used in early development to have the formulation of Table 20, Pabburi hydroxyethyl sulfonate. Both acid salt (ISE) capsules. After 10 minutes, the formulations A1, A2 and B were all superior to the tablets of Examples 1 to 7, and thus conformed to the solid dosage form dissolution target.

實施例20 Example 20 琥珀酸粒徑分布的衝擊 Impact of succinic acid particle size distribution

按收到的酸(至多1毫米直徑之寬粒徑分布)及酸的 三個篩出物(sieve cut)評定琥珀酸粒徑分布對琥珀醯基加成物形成率的衝擊。越小的酸粒徑導致越大的雜質形成率,因為其更大的比表面積能使酸與游離鹼API接觸的頻率更高。因此,以具有大於約100微米粒徑之酸在藥物產品調配中較佳,以改進化學穩定性。 According to the acid received (large particle size distribution of up to 1 mm diameter) and acid Three sieve cuts were used to determine the impact of the succinic acid particle size distribution on the amber-based adduct formation rate. The smaller the acid particle size results in a larger impurity formation rate because its larger specific surface area allows the acid to contact the free base API more frequently. Therefore, acids having a particle size greater than about 100 microns are preferred in pharmaceutical product formulation to improve chemical stability.

實施例21 Example 21 物理穩定性分析 Physical stability analysis

發展定量拉曼(Raman)光譜法來評估調配物的物理穩定性。拉曼方法係使用Kaiser Optical Systems PhAT探針且定量模式係利用自API、API琥珀酸鹽複合物及賦形劑所製備的一組校準標準物建立。調配物A1及B之相對穩定性係藉由測定錠劑在30至50℃及至多75% RH下貯存至多1個月之後在調配物中的API琥珀酸鹽複合物量來評定。 Quantitative Raman spectroscopy was developed to assess the physical stability of the formulation. The Raman method uses the Kaiser Optical Systems PhAT probe and the quantitative mode is established using a set of calibration standards prepared from API, API succinate complexes and excipients. The relative stability of formulations A1 and B was assessed by measuring the amount of API succinate complex in the formulation after storage of the tablet at 30 to 50 ° C and up to 75% RH for up to 1 month.

調配物B顯示相對於調配物A1而較低的琥珀酸鹽複合物轉換。轉換量係隨時間增加且以較高的溫度及濕度之貯存條件加速。 Formulation B showed a lower succinate complex conversion relative to formulation A1. The conversion amount increases with time and is accelerated by storage conditions of higher temperature and humidity.

以調配物A1及B在各種溫度及濕度條件下所形成的API琥珀酸鹽複合物量係以時間為函數顯示於表22中,其中〝LOD〞係指檢測限度及〝LOQ〞係指定量限度。 The amount of API succinate complex formed by formulations A1 and B under various temperature and humidity conditions is shown in Table 22 as a function of time, where 〝LOD〞 refers to the limit of detection and the specified amount of 〝LOQ 〞.

評定未包膜的原型錠劑調配物A1在開放條件下的物理穩定性。施予錠劑上升的溫度及濕度。使用拉曼光譜法在受應力樣品中觀察到琥珀酸鹽複合物之轉換。 The physical stability of the uncoated prototype tablet formulation A1 under open conditions was evaluated. The temperature and humidity at which the tablet is raised. Conversion of the succinate complex was observed in stressed samples using Raman spectroscopy.

比較調配物A1及B在40℃/75% RH下貯存1個月之後的化學純度。如表23中所示,調配物B顯示相對於調配物A1而顯著較低的總雜質形成程度。 The chemical purity of Formulations A1 and B after storage for 1 month at 40 ° C / 75% RH was compared. As shown in Table 23, Formulation B showed a significantly lower total impurity formation relative to Formulation A1.

以該等數據為基準測定濕度(%RH)對自游離鹼成為琥珀酸鹽複合物之API轉換率具有顯著的衝擊。增加溫度亦衝擊成為複合物的轉換,但是具有比濕度弱的影響。 Determination of humidity (%RH) based on these data has a significant impact on the API conversion rate from the free base to the succinate complex. Increasing the temperature also impacts the conversion of the composite, but has a weaker effect than the humidity.

在以金屬箔對金屬箔包裝(foil-foil packaging)之前在60% RH中預平衡的錠劑顯現快速的琥珀酸鹽複合物形成。第9週的穩定性數據表明需要控制在錠劑中的水活性,使藥物產品中的帕博西里琥珀酸鹽複合物形成減至最低。調配物B(其含有顆粒外硬脂基富馬酸鈉作為潤滑劑)顯示在高濕度(75%RH)條件下比調配物A1(其併入硬脂酸鎂作為顆粒外潤滑劑)低的琥珀酸鹽複合物轉換。 Tablets pre-equilibrated in 60% RH prior to metal foil-foil-foil packaging exhibited rapid succinate complex formation. The stability data for Week 9 indicated the need to control the water activity in the tablet to minimize the formation of the Pabsiri succinate complex in the drug product. Formulation B (which contains extragranular stearyl fumarate as a lubricant) shows lower than the formulation A1 (which incorporates magnesium stearate as an extragranular lubricant) under high humidity (75% RH) conditions. Succinate complex conversion.

Claims (20)

一種固態劑型,其包含從約10重量%至約35重量%之帕博西里、從約5重量%至約25重量%之選自由琥珀酸、蘋果酸和酒石酸所組成之群組的水溶性酸及醫藥上可接受之載劑。 A solid dosage form comprising from about 10% to about 35% by weight of Pabsiri, from about 5% to about 25% by weight of a water-soluble acid selected from the group consisting of succinic acid, malic acid and tartaric acid And a pharmaceutically acceptable carrier. 根據申請專利範圍第1項之固態劑型,其中該水溶性酸為琥珀酸。 A solid dosage form according to claim 1 wherein the water soluble acid is succinic acid. 根據申請專利範圍第1或2項之固態劑型,其中該醫藥上可接受之載劑包含至少一種稀釋劑,且其中該稀釋劑構成該固態劑型的約50重量%至約75重量%。 The solid dosage form according to claim 1 or 2, wherein the pharmaceutically acceptable carrier comprises at least one diluent, and wherein the diluent constitutes from about 50% to about 75% by weight of the solid dosage form. 根據申請專利範圍第3項之固態劑型,其中該稀釋劑係選自由下列所組成之群組:微晶纖維素、乳糖單水合物、甘露醇、山梨醇、木糖醇、碳酸鎂、磷酸氫鈣(dibasic calcium phosphate)和磷酸鈣(tribasic calcium phosphate)。 The solid dosage form according to claim 3, wherein the diluent is selected from the group consisting of microcrystalline cellulose, lactose monohydrate, mannitol, sorbitol, xylitol, magnesium carbonate, hydrogen phosphate Dibasic calcium phosphate and tribasic calcium phosphate. 根據申請專利範圍第1項之固態劑型,其中該醫藥上可接受之載劑包含至少一種潤滑劑,且其中該潤滑劑構成該固態劑型的從約0.5重量%至約10重量%。 The solid dosage form of claim 1, wherein the pharmaceutically acceptable carrier comprises at least one lubricant, and wherein the lubricant comprises from about 0.5% to about 10% by weight of the solid dosage form. 根據申請專利範圍第5項之固態劑型,其中該潤滑劑係選自由下列所組成之群組:硬脂酸鎂、硬脂酸鈣、硬脂酸鋅和硬脂基富馬酸鈉。 A solid dosage form according to claim 5, wherein the lubricant is selected from the group consisting of magnesium stearate, calcium stearate, zinc stearate and sodium stearyl fumarate. 根據申請專利範圍第1項之固態劑型,其中該醫藥上可接受之載劑包含至少一種崩解劑,且其中該崩解劑構成該固態劑型的從約5重量%至約10重量%。 The solid dosage form of claim 1, wherein the pharmaceutically acceptable carrier comprises at least one disintegrant, and wherein the disintegrant comprises from about 5% to about 10% by weight of the solid dosage form. 根據申請專利範圍第7項之固態劑型,其中該崩解劑係選自由下列所組成之群組:交聯聚維酮(crospovidone)、交聯羧甲基纖維素鈉(croscarmellose sodium)和乙醇酸澱粉鈉(sodium starch glycolate)。 A solid dosage form according to claim 7 wherein the disintegrant is selected from the group consisting of crospovidone, croscarmellose sodium and glycolic acid. Sodium starch glycolate. 根據申請專利範圍第1項之固態劑型,其係呈錠劑型式。 The solid dosage form according to item 1 of the patent application is in the form of a tablet. 根據申請專利範圍第9項之固態劑型,其中該錠劑經包膜。 A solid dosage form according to claim 9 wherein the tablet is coated. 根據申請專利範圍第9項之固態劑型,其中該錠劑為雙層錠劑。 A solid dosage form according to claim 9 wherein the tablet is a bilayer tablet. 根據申請專利範圍第2項之固態劑型,其中當該劑型添加至在具有於50rpm自旋之葉片的標準USP 2旋轉葉片裝置中包含37℃下500毫升10mM pH 5.5之乙酸鹽緩衝液的試驗介質中時,其溶解:(a)在15分鐘內不少於35%之帕博西里;(b)在30分鐘內不少於45%之帕博西里;(c)在60分鐘內不少於55%之帕博西里;或(d)(a)、(b)及(c)中之二或更多者。 The solid dosage form according to claim 2, wherein the dosage form is added to a test medium comprising 500 ml of 10 mM acetate buffer of pH 5.5 at 37 ° C in a standard USP 2 rotary vane apparatus having blades with spins at 50 rpm. In the middle, it dissolves: (a) not less than 35% of Pabori in 15 minutes; (b) not less than 45% of Pabori in 30 minutes; (c) not less than 60 minutes in 60 minutes 55% of Pabbori; or (d) two or more of (a), (b) and (c). 根據申請專利範圍第2項之固態劑型,其中當該劑型添加至在具有於50rpm自旋之葉片的標準USP 2旋轉葉片裝置中包含37℃下500毫升50mM pH 6.5之磷酸鹽緩衝液及0.1M NaCl的試驗介質中時,其溶解:(a)在15分鐘內不少於40%之帕博西里;(b)在30分鐘內不少於35%之帕博西里;(c)在60分鐘內不少於25%之帕博西里;或(d)(a)、(b)及(c)中之二或更多 者。 The solid dosage form according to claim 2, wherein the dosage form is added to a standard USP 2 rotary vane device having a blade having a spin of 50 rpm, comprising 500 ml of 50 mM phosphate buffer pH 6.5 at 37 ° C and 0.1 M In the test medium of NaCl, it dissolves: (a) not less than 40% of Pabori in 15 minutes; (b) less than 35% of Pabori in 30 minutes; (c) in 60 minutes Not less than 25% of Pabori; or (d) two or more of (a), (b) and (c) By. 根據申請專利範圍第2項之固態劑型,其中該劑型:(a)在投予受試者單一經口劑量之後在血漿濃度相對時間曲線下面積(AUC)具有從約0.8至約1.25之平均進食/空腹比;(b)在投予受試者單一經口劑量之後具有從約0.8至約1.25之最大血漿濃度(Cmax)的平均進食/空腹比;或(c)(a)與(b)二者。 The solid dosage form according to item 2 of the scope of the patent application, wherein the dosage form: (a) has an average area (AUC) of from about 0.8 to about 1.25 in a plasma concentration versus time curve after administration of a single oral dose to the subject. / fasting ratio; (b) an average eating/fasting ratio having a maximum plasma concentration ( Cmax ) from about 0.8 to about 1.25 after administration of a single oral dose to the subject; or (c) (a) and (b) )both. 根據申請專利範圍第2項之固態劑型,其中該劑型:(a)在投予受試者單一經口劑量之後提供含有等效量的帕博西里之對照用即時釋放型(IR)經口膠囊的80%至125%之平均空腹AUC的範圍內之平均空腹AUC;(b)在投予受試者單一經口劑量之後提供含有等效量的帕博西里之對照用即時釋放型(IR)經口膠囊的80%至125%之平均空腹Cmax的範圍內之平均空腹Cmax;或(c)(a)與(b)二者。 A solid dosage form according to item 2 of the scope of the patent application, wherein the dosage form: (a) provides a controlled immediate release (IR) oral capsule containing an equivalent amount of Pabsiri after administration of a single oral dose to the subject Average fasting AUC within the range of 80% to 125% of the average fasting AUC; (b) providing an immediate release type (IR) containing an equivalent amount of Pabsiri after administration of a single oral dose to the subject the mean 80-125% of the opening of the capsule fasting average of the range of fasting plasma C max C max; or (c) (a) and both (b). 根據申請專利範圍第2項之固態劑型,其中該劑型提供:(a)在投予受試者單一經口劑量之後在質子泵抑制劑(PPI)存在下的平均AUC係在沒有PPI存在下的平均AUC之80%至125%的範圍內;(b)在投予受試者單一經口劑量之後在質子泵抑制劑(PPI)存在下的平均Cmax係在沒有PPI存在下的平均Cmax之80%至125%的範圍內;或(c)(a)與(b)二者。 A solid dosage form according to claim 2, wherein the dosage form provides: (a) an average AUC line in the presence of a proton pump inhibitor (PPI) after administration of a single oral dose to the subject in the absence of PPI in the range of 80-125% of the mean AUC; (b) at a mean C max based proton pump inhibitor (PPI) is present in the subject after a single oral administration dose mean C max occurring in the absence of PPI 80% to 125% of the range; or (c) both (a) and (b). 根據申請專利範圍第16項之固態劑型,其中該PPI為瑞貝拉唑(rabeprazole)。 A solid dosage form according to claim 16 wherein the PPI is rabeprazole. 根據申請專利範圍第2項之固態劑型,其中該劑型在25℃及60% RH下貯存1年之後展現少於0.05重量%之酸加成物。 A solid dosage form according to claim 2, wherein the dosage form exhibits less than 0.05% by weight acid adduct after storage for one year at 25 ° C and 60% RH. 根據申請專利範圍第2項之固態劑型,其中在該劑型中的帕博西里之量為25毫克、75毫克、100毫克或125毫克。 A solid dosage form according to item 2 of the scope of the patent application, wherein the amount of paclitaxel in the dosage form is 25 mg, 75 mg, 100 mg or 125 mg. 根據申請專利範圍第19項之固態劑型,其中在該劑型中的帕博西里之量為125毫克。 A solid dosage form according to claim 19, wherein the amount of Pabbori in the dosage form is 125 mg.
TW105117213A 2015-06-04 2016-06-01 Solid dosage forms of palbociclib TWI635863B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562171177P 2015-06-04 2015-06-04
US62/171,177 2015-06-04
US201662332973P 2016-05-06 2016-05-06
US62/332,973 2016-05-06

Publications (2)

Publication Number Publication Date
TW201711687A true TW201711687A (en) 2017-04-01
TWI635863B TWI635863B (en) 2018-09-21

Family

ID=56092955

Family Applications (2)

Application Number Title Priority Date Filing Date
TW107123537A TWI763881B (en) 2015-06-04 2016-06-01 Solid dosage forms of palbociclib
TW105117213A TWI635863B (en) 2015-06-04 2016-06-01 Solid dosage forms of palbociclib

Family Applications Before (1)

Application Number Title Priority Date Filing Date
TW107123537A TWI763881B (en) 2015-06-04 2016-06-01 Solid dosage forms of palbociclib

Country Status (31)

Country Link
US (2) US11065250B2 (en)
EP (2) EP3302565B1 (en)
JP (3) JP2017002034A (en)
KR (2) KR102068423B1 (en)
CN (2) CN113616606A (en)
AU (2) AU2016272881C1 (en)
BR (1) BR112017025398A2 (en)
CA (1) CA2931892A1 (en)
CL (1) CL2017003089A1 (en)
CO (1) CO2017012362A2 (en)
CR (1) CR20170540A (en)
CY (1) CY1122454T1 (en)
DK (1) DK3302565T3 (en)
DO (1) DOP2017000280A (en)
ES (1) ES2764459T3 (en)
HK (1) HK1250570A1 (en)
HR (1) HRP20192065T1 (en)
HU (1) HUE047477T2 (en)
IL (2) IL255632A (en)
LT (1) LT3302565T (en)
MX (2) MX2017015579A (en)
PE (1) PE20180395A1 (en)
PL (1) PL3302565T3 (en)
PT (1) PT3302565T (en)
RS (1) RS59672B1 (en)
RU (1) RU2686840C1 (en)
SA (1) SA517390473B1 (en)
SI (1) SI3302565T1 (en)
TW (2) TWI763881B (en)
WO (1) WO2016193860A1 (en)
ZA (1) ZA201707780B (en)

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2350096T3 (en) 2008-10-02 2020-06-29 Salix Pharmaceuticals, Ltd. Methods of treating hepatic encephalopathy
CN105616418A (en) * 2014-11-07 2016-06-01 江苏豪森药业集团有限公司 Pharmaceutical preparation containing cyclin inhibitor, and preparation method thereof
WO2017115315A1 (en) * 2015-12-30 2017-07-06 Dr. Reddy's Laboratories Limited Solid forms of palbociclib
CN109078006B (en) * 2016-03-29 2021-04-20 深圳市药欣生物科技有限公司 Medicinal preparation of palbociclib and preparation method thereof
US10449195B2 (en) 2016-03-29 2019-10-22 Shenzhen Pharmacin Co., Ltd. Pharmaceutical formulation of palbociclib and a preparation method thereof
CN108210454A (en) * 2016-12-14 2018-06-29 深圳市华力康生物医药有限公司 A kind of Orally taken emulsion of Pabuk former times profit cloth and preparation method thereof
AU2018210141A1 (en) * 2017-01-20 2019-08-29 Constellation Pharmaceuticals, Inc. Solid dispersions of (R)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide
WO2018165404A1 (en) * 2017-03-08 2018-09-13 Cinrx Pharma, Llc Pharmaceutical formualtions of phloroglucinol and trimethylphloroglucinol
WO2018191950A1 (en) * 2017-04-21 2018-10-25 Alnova Pharmaceuticals, Ltd. Palbociclib compositions and methods thereof
US10603292B2 (en) * 2017-05-31 2020-03-31 Boehringer Ingelheim International Gmbh Pharmaceutical composition and pharmaceutical dosage form comprising (E)-4-(2-(aminomethyl)-3-fluoroallyloxy)-N-tert-butylbenzamide, process for their preparation, methods for treating and uses thereof
CN106970177B (en) * 2017-06-06 2018-05-15 北京元延医药科技股份有限公司 The analyzing detecting method of Pa Boxini intermediates and its impurity
SG11202000136YA (en) 2017-07-10 2020-02-27 Takeda Pharmaceuticals Co Preparation comprising vonoprazan
US11529353B2 (en) * 2017-07-28 2022-12-20 Synthon B.V. Pharmaceutical composition comprising Palbociclib
CN110997666A (en) * 2017-09-19 2020-04-10 浙江华海药业股份有限公司 N-formyl palbociclib, preparation method and application thereof, palbociclib preparation and quality control method thereof
TWI803530B (en) * 2017-10-27 2023-06-01 美商普雷辛肯公司 Formulations of a compound modulating kinases
CN108014343A (en) * 2017-12-21 2018-05-11 孟斯琴 A kind of pharmaceutical composition for treating breast cancer and preparation method thereof
CN108066303A (en) * 2017-12-31 2018-05-25 湖南博隽生物医药有限公司 A kind of anticancer pharmaceutical composition and preparation method thereof
WO2019166928A1 (en) 2018-02-27 2019-09-06 Pfizer Inc. Combination of a cyclin dependent kinase inhibitor and a bet-bromodomain inhibitor
JP2021523202A (en) * 2018-05-14 2021-09-02 ファイザー・インク Oral solution preparation
JP2021523882A (en) * 2018-05-15 2021-09-09 ジエンス ハンセン ファーマセウティカル グループ カンパニー リミテッド A pharmaceutical composition containing a small molecule EGFR inhibitor and a method for producing the same.
CN108653222A (en) * 2018-07-03 2018-10-16 威海贯标信息科技有限公司 A kind of Pa Boxini tablet compositions
CN109575021B (en) * 2019-01-22 2020-09-18 北京海美桐医药科技有限公司 Preparation method of thuja occidentalis
US20220125777A1 (en) 2019-02-01 2022-04-28 Pfizer Inc. Combination of a cdk inhibitor and a pim inhibitor
US20230044749A1 (en) 2019-12-16 2023-02-09 Lunella Biotech, Inc. Selective cdk 4/6 inhibitor cancer therapeutics
AU2020405427A1 (en) 2019-12-16 2022-07-14 Lunella Biotech, Inc. Selective CDK4/6 inhibitor cancer therapeutics
US20230364063A1 (en) * 2020-07-02 2023-11-16 Artham Therapeutics Inc. Oral pharmaceutical composition and method for producing same
WO2022018596A1 (en) 2020-07-20 2022-01-27 Pfizer Inc. Combination therapy
CN114246872B (en) * 2020-09-24 2024-02-06 南京济群医药科技股份有限公司 Technological method and composition for improving fluidity of isethionic acid piperazine Bai Xili
CN114306245A (en) 2020-09-29 2022-04-12 深圳市药欣生物科技有限公司 Pharmaceutical composition of amorphous solid dispersion and preparation method thereof
WO2022091001A1 (en) 2020-10-29 2022-05-05 Pfizer Ireland Pharmaceuticals Process for preparation of palbociclib
WO2022123419A1 (en) 2020-12-08 2022-06-16 Pfizer Inc. Treatment of luminal subtypes of hr-positive, her2-negative early breast cancer with palbociclib
WO2022162122A1 (en) 2021-01-29 2022-08-04 Biotx.Ai Gmbh Genetically verified netosis inhibitor for use in the treatment of a sars-cov2 infection
EP4302755A1 (en) 2022-07-07 2024-01-10 Lotus Pharmaceutical Co., Ltd. Palbociclib formulation containing an amino acid
EP4302832A1 (en) 2022-07-07 2024-01-10 Lotus Pharmaceutical Co., Ltd. Palbociclib formulation containing glucono delta lactone
WO2024132652A1 (en) 2022-12-22 2024-06-27 Synthon B.V. Pharmaceutical composition comprising palbociclib
WO2024133726A1 (en) 2022-12-22 2024-06-27 Synthon B.V. Pharmaceutical composition comprising palbociclib

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9518953D0 (en) 1995-09-15 1995-11-15 Pfizer Ltd Pharmaceutical formulations
TW477787B (en) 1996-08-27 2002-03-01 Pfizer Pyrido six-membered nitrogen-containing cyclic ring derivatives having corticotropin releasing factor antagonist activity and pharmaceutical composition containing same
EP0946554A1 (en) 1996-11-27 1999-10-06 Pfizer Inc. Fused bicyclic pyrimidine derivatives
KR20060111716A (en) 2002-01-22 2006-10-27 워너-램버트 캄파니 엘엘씨 2-(pyridin-2-ylamino)-pyrido[2,3-d]pyrimidin-7-ones
US20030181488A1 (en) 2002-03-07 2003-09-25 Boehringer Ingelheim Pharma Gmbh & Co. Kg Administration form for the oral application of 3-[(2-{[4-(hexyloxycarbonylamino-imino-methyl)-phenylamino]-methyl}-1-methyl-1H-benzimidazol-5-carbonyl)-pyridin-2-yl-amino]-propionic acid ethyl ester and the salts thereof
EP1648889B1 (en) 2003-07-11 2008-10-29 Warner-Lambert Company LLC Isethionate salt of a selective cdk4 inhibitor
GB0317663D0 (en) * 2003-07-29 2003-09-03 Astrazeneca Ab Pharmaceutical composition
BRPI0614870A2 (en) * 2005-08-22 2011-04-19 Novartis Ag pharmaceutical compositions
GB0517204D0 (en) * 2005-08-22 2005-09-28 Novartis Ag Organic compounds
CA2640382C (en) 2006-01-27 2015-12-29 Eurand, Inc Drug delivery systems comprising weakly basic drugs and organic acids
RU2009108006A (en) 2006-09-08 2010-10-20 Пфайзер Продактс Инк. (Us) SYNTHESIS OF 2- (PYRIDIN-2-ILAMINO) -PYRIDO [2, 3-D] PYRIMIDIN-7-ONES
KR101099926B1 (en) 2006-09-15 2011-12-28 화이자 프로덕츠 인코포레이티드 Pyrido 2,3-d pyrimidinone compounds and their use as pi3 inhibitors
WO2011039527A1 (en) * 2009-09-30 2011-04-07 Merck Sharp & Dohme Ltd Formulations for c-met kinase inhibitors
MX2012014387A (en) 2010-06-09 2013-05-01 Abbvie Inc Solid dispersions containing kinase inhibitors.
LT2782557T (en) 2011-11-23 2018-12-27 Array Biopharma, Inc. Pharmaceutical formulations
CA2860098A1 (en) 2011-12-22 2013-06-27 Boehringer Ingelheim International Gmbh Immediate release multi unit pellet system
DK2958916T3 (en) 2013-02-21 2018-11-12 Pfizer Solid forms of a selective CDK4 / 6 inhibitor
DK3033086T3 (en) 2013-08-14 2022-01-03 Novartis Ag Combination therapy for the treatment of cancer
KR20160084438A (en) 2013-11-13 2016-07-13 노파르티스 아게 Mtor inhibitors for enhancing the immune response
WO2016156070A1 (en) 2015-04-02 2016-10-06 Sandoz Ag Modified particles of palbociclib
US10449195B2 (en) 2016-03-29 2019-10-22 Shenzhen Pharmacin Co., Ltd. Pharmaceutical formulation of palbociclib and a preparation method thereof
CN109078006B (en) 2016-03-29 2021-04-20 深圳市药欣生物科技有限公司 Medicinal preparation of palbociclib and preparation method thereof

Also Published As

Publication number Publication date
PE20180395A1 (en) 2018-02-28
EP3302565B1 (en) 2019-11-06
DK3302565T3 (en) 2020-01-02
ZA201707780B (en) 2020-05-27
AU2016272881C1 (en) 2019-10-03
CY1122454T1 (en) 2021-01-27
IL255632A (en) 2018-01-31
JP2017002034A (en) 2017-01-05
JP2021167343A (en) 2021-10-21
KR102369405B1 (en) 2022-03-02
PL3302565T3 (en) 2020-06-01
TW201906611A (en) 2019-02-16
HUE047477T2 (en) 2020-04-28
RS59672B1 (en) 2020-01-31
KR20200006633A (en) 2020-01-20
MX2020003825A (en) 2020-08-06
CN107666914A (en) 2018-02-06
CN113616606A (en) 2021-11-09
IL287437A (en) 2021-12-01
CL2017003089A1 (en) 2018-05-11
TWI635863B (en) 2018-09-21
TWI763881B (en) 2022-05-11
DOP2017000280A (en) 2018-01-15
AU2019204689B2 (en) 2020-12-03
CO2017012362A2 (en) 2018-03-28
RU2686840C1 (en) 2019-05-06
CR20170540A (en) 2018-02-02
AU2016272881A1 (en) 2017-12-07
LT3302565T (en) 2019-12-27
JP2023112149A (en) 2023-08-10
NZ737391A (en) 2023-11-24
AU2016272881B2 (en) 2019-04-11
KR20180015232A (en) 2018-02-12
HRP20192065T1 (en) 2020-02-21
EP3636283A1 (en) 2020-04-15
US20180207100A1 (en) 2018-07-26
WO2016193860A1 (en) 2016-12-08
MX2017015579A (en) 2018-03-27
KR102068423B1 (en) 2020-01-20
US20210315900A1 (en) 2021-10-14
CA2931892A1 (en) 2016-12-04
US11065250B2 (en) 2021-07-20
BR112017025398A2 (en) 2018-08-07
PT3302565T (en) 2020-01-14
SI3302565T1 (en) 2020-02-28
AU2019204689A1 (en) 2019-07-18
HK1250570A1 (en) 2018-12-28
ES2764459T3 (en) 2020-06-03
SA517390473B1 (en) 2022-12-11
EP3302565A1 (en) 2018-04-11

Similar Documents

Publication Publication Date Title
AU2019204689B2 (en) Solid dosage forms of palbociclib
TWI778983B (en) Tablets comprising 2-hydroxy-6-((2-(1-isopropyl-1h-pyrazol-5-yl)pyridin-3-yl)methoxy)benzaldehyde
JP6404217B2 (en) Enzalutamide formulation
JP5026635B2 (en) Rapid release pharmaceutical composition of pharmaceutical substance
RU2351316C2 (en) Dosage forms with retarded release of ziprasidone
US20080167362A1 (en) Celecoxib compositions
JP2009542647A (en) Memantine pharmaceutical composition
EP0514967A1 (en) Low solubility drug-coated bead compositions
WO2011001202A1 (en) Solid pharmaceutical fixed dose compositions comprising irbesartan and amlodipine, their preparation and their therapeutic application
JP2023052494A (en) Instant release pharmaceutical preparation of anticoagulant and preparation method therefor
WO2017093890A1 (en) Clobazam tablet formulation and process for its preparation
KR20040047920A (en) Solid pharmaceutical formulation for a piperazine urea derivative
WO2018130943A1 (en) Oral pharmaceutical composition of lurasidone and preparation thereof
KR102136459B1 (en) Combination formulation comprising bazedoxifene and vitamin D